Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 Mar 23.
Published in final edited form as: Curr Neurovasc Res. 2010 Feb 1;7(1):59–64. doi: 10.2174/156720210790820217

Diabetes Mellitus: Channeling Care through Cellular Discovery

Kenneth Maiese 1,2,3,4,5,*, Yan Chen Shang 1, Zhao Zhong Chong 1, Jinling Hou 1
PMCID: PMC2844489  NIHMSID: NIHMS183028  PMID: 20158461

Abstract

Diabetes mellitus (DM) impacts a significant portion of the world’s population and care for this disorder places an economic burden on the gross domestic product for any particular country. Furthermore, both Type 1 and Type 2 DM are becoming increasingly prevalent and there is increased incidence of impaired glucose tolerance in the young. The complications of DM are protean and can involve multiple systems throughout the body that are susceptible to the detrimental effects of oxidative stress and apoptotic cell injury. For these reasons, innovative strategies are necessary for the implementation of new treatments for DM that are generated through the further understanding of cellular pathways that govern the pathological consequences of DM. In particular, both the precursor for the coenzyme β-nicotinamide adenine dinucleotide (NAD+), nicotinamide, and the growth factor erythropoietin offer novel platforms for drug discovery that involve cellular metabolic homeostasis and inflammatory cell control. Interestingly, these agents and their tightly associated pathways that consist of cell cycle regulation, protein kinase B, forkhead transcription factors, and Wnt signaling also function in a broader sense as biomarkers for disease onset and progression.

Keywords: Aging, biomarkers, diabetes, erythropoietin, FoxO3a, oxidative stress, sirtuins, Wnt

DIABETES MELLITUS, ECONOMIC BURDEN, AND MULTI-SYSTEM COMPLICATIONS

Diabetes mellitus (DM) is a metabolic disorder closely associated with increased weight gain [1, 2] and affects almost 20 million individuals in the United States and more than 165 million individuals worldwide [3]. By 2030, DM may be a condition to contend with in more than 360 million individuals. Furthermore, a significant portion of the population has undiagnosed diabetes, illustrating the need for improved early diagnosis [4]. Type 1 insulin-dependent DM is present in 5–10 percent of all diabetics, but is increasing in adolescent minority groups [1, 5]. Type 1 DM also leads to long-term complications throughout the body involving cardiovascular, renal, and nervous system disease [6]. Type 2 DM represents at least 80 percent of all diabetics, usually in individuals over 40 years of age, and is dramatically increasing in incidence as a result of changes in human behavior and increased body mass index [1, 5]. Type 2 DM is characterized by a progressive deterioration of glucose tolerance with early β-cell compensation for insulin resistance (achieved by β-cell hyperplasia). This is subsequently followed by progressive decrease in β-cells mass. In addition, the growing incidence of impaired glucose tolerance in the young also is worrisome [7].

These statistics bring to light the emerging burden DM presents to the world healthcare system. Compared with other nations, the United States devotes 16% of the gross domestic product to healthcare. In addition, spending for each individual is the highest level in the world and is equal to $7,290 [8]. Total spending on pharmaceuticals is also the highest in the world with $878 per individual. However, life expectancy in years in the United States equals 78.1 years and trails behind other developed countries such as Japan (life expectancy of 82.6 years). The United States also is ranked as having the highest level of obesity in the population at 34.3% while countries such as Japan have a 3.4% level of obesity [8]. Patients with DM can develop a number of disorders that include immune dysfunction [9], sarcopenia [10], depression [11], hepatic dysfunction [12], renal disease [13], anemia and hematological disease [1416], neurodegenerative disorders [5, 17, 18], and cardiovascular disease [5, 19]. Interestingly, patients with DM are at risk for the development of cognitive disorders that may be associated with vascular disease [9, 20]. In relation to neurodegenerative disease, DM may be associated with the development of Alzheimer’s disease [21, 22]. Alzheimer’s disease can be the result of a number of etiologies [23, 24], such as changes in cerebral blood flow and metabolism with aging [25], sialylation and glycosylation of amyloid plaques [26, 27], aberrant cell cycle induction [2830], amyloid toxicity [3034], chemokine induction [35], exogenous toxins [36], alteration in muscarinic and nicotinic pathways [25, 37], and intracellular calcium changes [38]. Additional studies for cognitive loss point to metabolic dysfunction [21, 39, 40]. For example, in animal models with brain/neuronal insulin receptor knockouts, loss of insulin signaling appears to be linked to increased phosphorylation of the microtubule-associated protein tau that occurs during Alzheimer’s disease [41]. In clinical investigations, some studies report that diabetic patients may have significantly less neuritic plaques and neurofibrillary tangles than non-diabetic patients [42], but other investigators report a modest adjusted relative risk of Alzheimer’s disease in patients with diabetes as compared with those without diabetes to be 1.3 [43]. Additional studies have described the reduced expression of genes encoding insulin in Alzheimer’s patients that suggests a potential link between DM and the development of Alzheimer’s disease [44].

DIABETES MELLITUS, OXIDANT PATHWAYS, AND INFLAMMATORY CELLS

These complications of DM are the result of multiple factors, but argue for the implementation of novel drug development strategies that are generated through the further understanding of cellular pathways that govern the pathological consequences of DM. In particular, cellular pathways that lead to DM and its complications have been tied to oxidant stress [1, 2, 17, 18, 21, 45, 46]. In studies with Type 1 diabetic animals, oxidative stress leads to DNA damage in renal cortical cells [47]. Although early effects of elevated glucose may increase the presence of potentially protective pathways [48], more prolonged exposure of elevated glucose [15, 49] as was as the rise in insulin levels [50] can lead to reactive oxygen species (ROS) and can be detrimental even if glucose levels are controlled [51]. In addition, elevated levels of ceruloplasmin during hyperglycemia are suggestive of increased ROS [52]. A number of treatment entities seek to ameliorate the effects of oxidative stress during DM [34, 5356].

Oxidative stress also may promote the onset of DM by decreasing insulin sensitivity and destroying the insulin-producing cells. ROS can penetrate through cell membranes and cause damage to β-cells of pancreas [57, 58]. A high fat diet [59] or free fatty acids also have been shown to release ROS and contribute to mitochondrial DNA damage and impaired pancreatic β-cell function [60]. In non-diabetic rats, hyperglycemia has been shown to increase muscle protein carbonyl content and elevated levels of malondialdehyde and 4-hydroxynonenal, indicators of oxidative stress and lipid peroxidation [61]. These biomarkers of oxidative stress and insulin resistance suggest that ROS contribute to the pathogenesis of hyperglycemia-induced insulin resistance [62, 63]. Hyperglycemia can lead to increased production of ROS in several cell types, such as pancreatic beta-cells and vascular cells [63, 64]. Chronic hyperglycemia is not necessary to lead to oxidative stress injury, since even short periods of hyperglycemia, generate ROS, such as in vascular cells [65]. Recent clinical correlates support these experimental studies to show that acute glucose swings in addition to chronic hyperglycemia can trigger oxidative stress mechanisms during Type 2 DM, demonstrating the importance for therapeutic interventions during acute and sustained hyperglycemic episodes [66].

At the cellular level, ROS are oxygen free radicals and other chemical entities that may lead to cell injury [28, 67, 68], but also have been tied to pain sensitivity [69]. Oxidative stress can result in hepatic injury [70, 71], pancreatitis [72], impaired cognition and psychiatric disorders [22, 73, 74], neuronal injury [23, 28, 7579], Parkinson’s disease [8083], complications of epilepsy [84], cardiovascular disease [8587], ocular disease [88], age-related disorders [89], metal ion injury [90], and promote xenobiotic toxicity [91, 92]. ROS consist of superoxide free radicals, hydrogen peroxide, singlet oxygen, nitric oxide (NO), and peroxynitrite [17, 23, 64]. ROS usually occur at low levels during normal physiological conditions and are scavenged by endogenous antioxidant systems that include superoxide dismutase (SOD), glutathione peroxidase, catalase, and vitamin D3 [93, 94]. Additional pathways include vitamins C, E, and K [84, 9598].

Oxidative stress leads to both apoptotic and non-apop-totic pathways. Apoptosis can occur during DM [1, 2, 99, 100], anesthetic exposure [101], tissue ischemia [102105], bone fatigue [106], neurodegenerative disorders [17, 107109] and Alzheimer’s disease [3033, 38, 110115], plasticity associated with ischemic preconditioning [116], aging-related diseases [18, 117, 118], and toxic conditions during development [101, 119]. During apoptosis, the cleavage of genomic DNA into fragments [108, 120, 121] usually occurs after the exposure of membrane phosphatidylserine (PS) residues [122127]. Membrane PS exposure occurs in neurons, vascular cells, and inflammatory microglia during reduced oxygen exposure [29, 68, 120, 128, 129], β-amyloid (Aβ) exposure [32, 34], nitric oxide exposure [130134], and during the administration of agents that induce the production of reactive oxygen species (ROS), such as 6-hydroxydopamine [83]. Membrane PS externalization also occurs on platelets and has been associated with clot formation in the vascular system [135].

The activation of inflammatory cells such as microglia that can dispose of injured cells is also dependent upon membrane PS externalization [136, 137]. For their own survival, microglia and non-neuronal cells of the brain are dependent upon several intracellular pathways, such as mTOR [138, 139] and zinc regulation [140]. Non-neuronal cells of the brain can be beneficial to modulate neurogenesis [141], to function as immune surveillance for toxic products [142], such as for β-amyloid [27], to block foreign organisms and viral agents from proliferating in the brain [143], to modulate vascular growth [144], and to allow for the repair of tissues composed of neuronal and vascular cells [138, 145]. However, microglia once stimulated also can promote cell injury and death. Microglia generate ROS [146, 147] that can worsen events with oxidative stress injury [148] and activate cytokines that in some circumstances may initially lead to cell proliferation [149], but later can result in the demise of cells [35, 143, 144]. For these reasons, it is important to understand the mechanisms that can activate microglia. Membrane PS exposure can become a signal for microglia to dispose of injured cells [40, 122, 125, 126, 150]. This process can be controlled by caspase 1 and caspase 3 [68, 151, 152]. Increased expression of the phosphatidylserine receptor (PSR) on microglia also occurs to facilitate activation of microglia [32, 56, 152, 153].

DIABETES MELLITUS, ENERGY MAINTENANCE, AND NICOTINAMIDE

Cellular pathways in DM are closely associated to cellular energy maintenance and intact mitochondrial function [5, 9, 45, 154156]. Oxidant stress and ROS exposure can result in the opening of the mitochondrial membrane permeability transition pore [125, 157159], reduce mitochondrial NAD+ stores [21], and result in apoptotic cell injury [23]. Free fatty acids also can lead to ROS release, mitochondrial DNA damage, and impaired pancreatic β-cell function [60]. In DM, mitochondrial dysfunctional also has been described. In patients with Type 2 DM, skeletal muscle mitochondria have been observed to be smaller than those in control subjects [160]. The development of Type 2 DM has been associated with a decrease in the levels of mitochondrial proteins and mitochondrial DNA [161].

One agent, namely nicotinamide, and the cellular pathways that it modulates may be critical for the maintenance of cellular energy homeostasis. Nicotinamide, the amide form of vitamin B3 (niacin) and nicotinamide or nicotinic acid is the water soluble form vitamin B3, is the principal form of niacin in dietary plant sources is nicotinic acid that is rapidly absorbed through the gastrointestinal epithelium [162]. Nicotinamide is subsequently generated through the conversion of nicotinic acid in the liver or through the hydrolysis of NAD+ [21]. Once nicotinamide is absorbed, it functions as the precursor for the coenzyme β-nicotinamide adenine dinucleotide (NAD+) [40, 163] and also is essential for the synthesis of nicotinamide adenine dinucleotide phosphate (NADP+) [164]. Nicotinamide is changed to its mononucleotide form (NMN) with the enzyme nicotinic acid/nicotinamide adenylyltransferase yielding the dinucleotides NAAD+ and NAD+. NAAD+ also yields NAD+ through NAD+ synthase [165] or NAD+ can be synthesized through nicotinamide riboside kinase that phosphorylates nicotinamide riboside to NMN [166, 167]. Nicotinamide through NAD+ is vital in cellular metabolism and can be directly utilized by cells to synthesize NAD+ [17, 40, 95]. Nicotinamide also participates in energy metabolism through the tricarboxylic acid cycle by utilizing NAD+ in the mitochondrial respiratory electron transport chain for the production of ATP, DNA synthesis, and DNA repair [168170].

During a number of conditions, nicotinamide is important for cell development and protection. Nicotinamide can be essential for the differentiation of human embryonic stem cells [171]. Nicotinamide also has more protean endocrine effects [172, 173] and offers protection for both neuronal [174176] and vascular cells [17, 40, 95, 163]. In neuronal cell populations, nicotinamide protects against free radical injury [159], anoxia [177], excitotoxicity [178], homocysteine toxicity [179], ethanol-induced neuronal injury [180], and oxygen-glucose deprivation [175, 181]. In cortical neurons, nicotinamide blocks cell injury during ROS generating toxins such as tertiary butylhydroperoxide [182]. Nicotinamide also can protect both rod and cone photoreceptor cells against N-methyl-N-nitrosourea toxicity [183, 184] as well as against glycation end products in all layers of the retina [185]. In animal studies, nicotinamide improves cognitive function, cell survival, and reduces edema following cortical trauma [186191], limits axonal degeneration [192], reduces cerebral ischemia [193195] sometimes more effectively in models that were absent of comorbidities [196], prevents spinal cord injury [197, 198], and lessens disability in models of Parkinson’s disease [80, 199, 200]. In vascular cells [174176], nicotinamide supports endothelial cell survival [40, 95, 163] especially during ROS exposure [159, 177, 201, 202] which may be crucial for tissue growth and repair [203]. Nicotinamide can protect the function of the blood brain barrier [186, 187], influence arteriolar dilatation and blood flow [204], increase skin vascular permeability [205], inhibit atherosclerotic plaque formation through inhibition of poly(ADP-ribose) polymerase [206], and foster platelet production through megakaryocyte maturation [207].

Interestingly, nicotinamide also may reverse a previously sustained early apoptotic injury [40, 159, 175, 177, 201, 208], suggesting that apoptosis prior to reaching genomic DNA degradation is dynamic and reversible in nature [40, 159, 177, 209]. Yet, some studies suggest that nicotinamide may either prevent or contribute to atherosclerotic plaques over a three to six month progression [210]. Although the mechanisms are not clear for the promotion of atherosclerosis, it is conceivable that these events may occur during oxidative stress and the production of acidosis-induced cellular toxicity [211213], since nicotinamide cannot prevent cellular injury during intracellular acidification paradigms [159].

More importantly, nicotinamide plays a significant role during DM. Nicotinamide has been suggested to have a close relationship with metabolic pathways that may lead to clinical cognitive changes [214]. Nicotinamide can maintain normal fasting blood glucose with streptozotocin-induced DM in animal models [215, 216], limit peripheral nerve injury during elevated glucose [217], reverse Type 1 DM in mice with acetyl-l-carnitine [218], and block oxidative stress [180, 201, 208, 219, 220]. Nicotinamide also affects levels of O-N-acetylglucosamin(O-GlcNAc)ylated proteins [221] and can significantly improve glucose utilization, prevent excessive lactate production in ischemic animal models [222]. In patients, nicotinamide (1200 mg/m2/day) protects β-cell function and prevents clinical disease in islet-cell antibody-positive first-degree relatives of Type 1 DM [223]. Nicotinamide also can reduce HbA1c levels in patients with recent onset Type 1 DM combined with intensive insulin therapy for up to two years after diagnosis [224]. Also relevant to patients with DM and renal insufficiency, nicotinamide can reduce intestinal absorption of phosphate and prevent the development of hyperphosphatemia [225]. Yet, some caution must be exercised with nicotinamide. Investigations have reported that elevated nicotinamide levels actually may foster DM [226] and that prolonged exposure to nicotinamide may lead to impaired β-cell function and reduction in cell growth [227, 228]. Furthermore, nicotinamide also may inhibit P450 and hepatic metabolism [229] and play a role in the progression of other disorders such as Parkinson’s disease [200].

There are several pathways that nicotinamide may use to maintain cellular metabolic homeostasis [18, 21, 40, 95, 171] and provide protection for cells during DM [159, 201]. Nicotinamide can function at the level of mitochondrial membrane pore formation [40, 201, 209] to prevent the release of cytochrome c [208] that may involve the phosphorylation of Bad [208]. Nicotinamide also can prevent mitochondrial membrane depolarization during exposure to either tert-butylhydroperoxide or atractyloside [175] and may inhibit the assembly of the mitochondrial permeability transition pore complex similar to the action of cyclosporin A [230] as well as stabilize cellular energy metabolism through ATP pathways [231]. Nicotinamide also blocks forkhead transcription factor activity, such as FoxO3a, through phosphorylation [175, 232], and may be protective through mechanisms of post-translational modification of FoxO3a and inhibition of caspase 3 activity [18, 137, 233235].

Nicotinamide also is closely linked to cell longevity pathways that involve sirtuins [95, 236, 237]. These include the sirtuin Sirt1, a NAD+-dependent deacetylase and the mammalian ortholog of the silent information regulator 2 (Sir2) protein [238], that can control multiple processes such as cell injury, lifespan, and metabolism [239, 240]. Sirtuins have been associated with cell longevity and aging as shown by early studies linking DAF-16 in Caenorhabditis elegans [137, 238, 240242]. Furthermore, sirtuins are tied to cellular metabolism [239, 243] and increased cell survival [240, 241, 244246]. However, the relationship between nicotinamide and sirtuins is not entirely clear, but sirtuin activation may promote glucose homeostasis and insulin sensitivity [21, 239, 240, 246, 247] while also reducing the risk of obesity [248]. In regards to nicotinamide, this agent prevents oxidative stress-induced apoptotic injury usually in a specific concentration range. Administration of nicotinamide in a range of 5.0–25.0 mmol/L significantly protects cells during oxidative stress injuries. This concentration range is similar to other injury paradigms in both animal models [184] and in cell culture models [40, 159, 201]. In contrast to these cytoprotective concentrations of nicotinamide that also can modulate gene regulation [249], a reduction in nicotinamide levels during nicotinamidase expression supports increased cellular survival and longevity [244, 246]. Nicotinamide can block cellular Sir2 by intercepting an ADP-ribosyl-enzyme-acetyl peptide intermediate with the regeneration of NAD+ (transglycosidation) [250]. Physiological concentrations of nicotinamide noncompetitively inhibit Sir2, promoting the premise that nicotinamide is a physiologically relevant regulator of Sir2 enzymes [251]. Yet, nicotinamidase expression, which reduces nicotinamide concentrations, prevents both apoptotic late DNA degradation and early PS exposure that appears to depend upon increased Sirt1 activity and may serve to modulate inflammatory cell activation [244, 246]. In addition, inhibition of sirtuin (Sirt1) activity either by pharmacological methods or siRNA gene silencing is detrimental to cell survival during oxidative stress and blocks nicotinamidase protection, further supporting that Sirt1 activity may be necessary for nicotinamidase protection during oxidative stress. As a result, it is the lower concentrations of nicotinamide that can function as an inhibitor of sirtuins that are necessary for the promotion of increased lifespan and cellular survival [175, 177, 201, 208, 244, 246, 252], at least in yeast and metazoans [95, 236, 237]. Furthermore, sirtuin activity also may prevent nicotinamide from assisting with DNA repair by altering the accessibility of DNA damaged sites for repair enzymes [253].

DIABETES MELLITUS, ERYTHROPOIETIN, AKT, FORKHEAD, AND WNT

Erythropoietin (EPO) is approved by the Food and Drug Administration for the treatment of anemia [63, 64]. Yet, enthusiasm for the use of this agent throughout the body has grown considerably since a growing number of investigations have identified EPO as an agent with effective utility for multiple disease processes [254, 255]. Treatment considerations include therapy for depression [256], Alzheimer’s disease [31, 257, 258], Parkinson’s disease [259], immune system dysfunction [130, 260263], neurodegeneration [31, 53, 130, 260, 264267], cardiovascular disorders [158, 261, 268276], spinal cord injury [277, 278], brain edema [279], fertility [280], trauma [281283], shock [284286], infection [287289], pulmonary disease [290292], renal disease [49, 293295], gastrointestinal disorders [296298], retinal disease and glaucoma [299302], and metabolic disorders [1, 16, 17, 53, 54, 303].

EPO is present in the brain, heart, and vascular system [260, 261, 270, 304306] as well as being required for erythropoiesis [307309]. EPO production occurs throughout the body [64, 255, 310] and can be detected in the breath of healthy individuals [311]. The principal organs of EPO production and secretion are the kidney, liver, brain, and uterus [232, 312, 313].

In patients with DM, plasma EPO is often low in individuals with anemia [314] or without anemia [315]. The inability of these individuals to produce EPO in response to declining hemoglobin levels may indicate an impaired EPO response during DM [316]. Yet, increased EPO secretion during diabetic pregnancies may represent the body’s attempt at endogenous protection against the complications of DM [317, 318]. EPO in diabetic as well as non-diabetic patients with severe, resistant congestive heart failure can decrease fatigue, increase left ventricular ejection fraction, and significantly decrease hospitalization stay [319]. In addition, EPO can serve to reverse the complications of anemia during DM [16]. The potential cytoprotective capacity of EPO may be important during other complications of DM, such as those that involve cognitive and neuronal synaptic impairment. Elevated EPO concentrations during infant maturation have been correlated with increased Mental Development Index scores [320] and EPO may prevent toxic effects of agents used to control cognitive function such as haloperidol [321]. In animal models, EPO may reduce apoptotic pathways during periods of hyperoxia in the developing brain [322, 323]. Furthermore, some clinical disorders may present with periods of hyperoxia followed by combined cerebral hypoperfusion and hypoxia that can lead to cerebral injury with associated oxidative stress [324]. EPO under these conditions may be protective since it can promote neurite outgrowth [325] and also may regulate hemoglobin levels that have recently been associated with cognitive decline [326]. Experimental work during elevated glucose also has demonstrated that EPO can significantly improve vascular cell survival in a 1.0 ng/ml range [54]. EPO administration in patients also can significantly increase plasma levels of EPO well above this range of 1.0 ng/ml that has been associated with potential EPO cellular protection in patients with cardiac or renal disease [327, 328], illustrating that the effects of EPO observed during in vitro studies may parallel the cellular processes altered by EPO in patients with DM [320].

Cellular protection by EPO is dependent upon multiple pathways, but three pathways in particular offer exciting perspectives for the future applications of EPO in clinical disease and especially during DM. One novel pathway involves protein kinase B (Akt). Activation of Akt can foster cell survival, such as during cell proliferation [329], progenitor cell development [149], blood-brain barrier permeability [330], inflammation [143, 331], ischemic-preconditioning [332], neurodegeneration [83], hyperglycemia [48, 333], hypoxia [270], amyloid toxicity [31, 32, 110, 111, 334], excitotoxicity [75], amyloid production [335], cardiomyopathy [336], cellular aging [337], and oxidative stress [125, 126, 338]. Akt activation also can modulate microglial cell activation [125, 126, 158], regulate transcription factors [261], maintain mitochondrial membrane potential (ΔΨm), prevent cytochrome c release [130, 158, 260], and block caspase activity [158, 260, 270]. The benefits of Akt to foster cell survival should be noted as potentially detrimental during tumor growth and during cancer resistance to chemotherapy [339].

EPO administration leads to the activation of Akt and protects against genomic DNA degradation and membrane PS exposure [130, 152, 260, 261, 270, 306, 340345]. Up-regulation of Akt activity, such as during vascular and cardiomyocyte ischemia [341, 342], free radical exposure [260, 346], matrix detachment [347], neuronal axotomy [348], N-methyl-D-aspartate toxicity [349], hypoxia [270, 350], β-amyloid toxicity [31, 351], DNA damage [126, 270, 338, 352], metabotropic receptor signaling [76, 85, 151, 353], cell metabolic pathways [40, 208], cytochrome c release [130, 270, 341], caspase activity [158, 260, 270], and oxidative stress [125, 126, 338] increases cell survival. Akt also can directly control microglial activation through the prevention of Bcl-xL degradation [338] and the inhibition of caspase 1-, 3-, and 9-like activities [125, 126, 151].

Akt also functions as a primary regulatory element for FoxOs, the forkhead transcription factors of the “O” class [238, 354]. These transcription factors bind to DNA through the forkhead domain that relies upon fourteen protein-DNA contacts [234, 354357]. The original nomenclature for these proteins, such as forkhead in rhabdomyosarcoma (FKHR), the Drosophila gene fork head (fkh), and Forkhead RElated ACtivator (FREAC)-1 and -2, has been replaced [233]. The present nomenclature for human Fox proteins places all letters in uppercase, otherwise only the initial letter is listed as uppercase for the mouse, and for all other chordates the initial and subclass letters are in uppercase [358]. Members of this family that include FoxO1, FoxO3, FoxO4, and FoxO6 are found throughout the body [63, 233, 235]. These proteins are expressed in tissues of the reproductive system of males and females, skeletal muscle, the cardiovascular system, lung, liver, pancreas, spleen, thymus, and the nervous system [137, 234, 357, 359]. Modulation of FoxOs is a viable therapeutic target for systems that involve metabotropic glutamate receptors [76], neurotrophins [360], cancer [137, 357, 361], and cytokines [261] to foster intended cell survival.

EPO modulates FoxOs for both cellular development and survival. EPO fosters eythroid progenitor cell development through the regulation of FoxO activity [255, 310, 362] and may require regulation of specific gene expression through an EPO-FoxO3a association to promote erythropoiesis in cultured cells [363]. In addition, EPO controls the phosphorylation and degradation of FoxO3a to retain it in the cytoplasm by binding to 14-3-3 protein and leads to increased cellular survival during oxidative stress [261]. Without the presence of EPO during oxidative stress injuries, FoxO3a is able to translocate from the cytoplasm of cells to the nucleus to initiate a “pro-apoptotic” program (Fig. 1).

Fig. (1). Subcellular trafficking of FoxO3a occurs during oxidative stress to allow for “pro-apoptotic” transcription.

Fig. (1)

Inflammatory microglial cells were imaged with immunofluorescent staining for FoxO3a (Texas-red streptavidin) six hours following oxidative stress exposure with oxygen-glucose deprivation (OGD). Nuclei of microglia were counterstained with DAPI. In merged images, untreated control microglia have readily visible nuclei (white/blue in color) that illustrate absence of FoxO3a in the nucleus. In contrast, merged images after OGD exposure show diffusely red cytoplasm and nucleus with minimal visibility of the nucleus with DAPI illustrating translocation of FoxO3a to the nucleus.

Yet, FoxO proteins are involved in a number of other cellular pathways that are also closely tied to DM. For example, FoxOs control cell cycle progression to prevent tumor growth [137, 357, 364]. Since attempted initiation of the cell cycle such as in neurons may be detrimental and can lead to cell death [28, 29, 365, 366], one may consider the ability of FoxO proteins to block cell cycle progression to be beneficial in these circumstances. In regards to DM, FoxO proteins may be cytoprotective. Interferon-gamma driven expression of tryptophan catabolism by cytotoxic T lymphocyte antigen 4 may activate Foxo3a to protect dendritic cells from injury in nonobese diabetic mice [367]. In addition, adipose tissue-specific expression of Foxo1 in mice improves glucose tolerance and sensitivity to insulin during an elevated fat diet [368]. FoxO proteins also may protect against diminished mitochondrial energy levels known to occur during insulin resistance such as in the elderly populations [1, 2, 5]. In caloric restricted mice that have decreased energy reserves, Foxo1, Foxo3a, and Foxo4 mRNA levels were noted to progressively increase over two years [369]. These observations complement studies in Drosophila and mammalian cells that demonstrate an increase in insulin signaling to regulate cellular metabolism during the up-regulation of FoxO1 expression [370].

It should be noted that the role of FoxO proteins in different cell systems can be variable and does not always point to a beneficial effect of FoxO proteins. FoxO3a controls early activation and subsequent apoptotic injury in microglia through caspase action of caspase 3, 8, and 9 [34, 56], illustrating that targeting FoxO3a activity may limit apoptotic caspase activity and promote cell survival. Analysis of the genetic variance in FOXO1a and FOXO3a on metabolic profiles, age-related diseases, fertility, fecundity, and mortality in patients have shown higher HbA1c levels and increased mortality risk associated with specific haplotypes of FOXO1a [371]. These clinical observations may indicate that elevated glucose levels can reduce post-translational phosphorylation of FOXO1, FOXO3a, and FOXO4 and initiate cellular apoptosis [372]. In addition, mice with a constitutively active Foxo1 transgene have increased microsomal triglyceride transfer protein and high plasma triglyceride levels [373]. Increased transcriptional activity of FoxO1, such as by the Sirt1 activator resveratrol, also can decrease insulin mediated glucose uptake and result in insulin resistance [374]. Overexpression of Foxo1 in skeletal muscles of mice can lead to a reduction in skeletal muscle mass and poor glycemic control [375]. Other studies that block the expression of Foxo1 in normal and cachectic mice [376] or reduce FoxO3 expression [377] demonstrate positive effects with an increase in skeletal muscle mass or resistance to muscle atrophy.

EPO also is intimately involved with a third pathway, namely those cell pathways controlled by Wnt signaling [17, 254, 378]. Wnt proteins derived from the Drosophila Wingless (Wg) and the mouse Int-1 genes are secreted cysteine-rich glycosylated proteins that play a role in a variety of cellular functions [32, 54, 153, 379]. Wnt proteins are involved in embryonic cell proliferation, cell differentiation, and cell survival that involve neurons, cardiomyocytes, endothelial cells, red blood cells, tumors, adipose tissue as well as several other cell types [235, 378, 380391].

In relation to metabolic conditions, abnormalities in Wnt pathways that involve transcription factor 7-like 2 gene may impart increased risk for type 2 DM in some populations [392394] and have a strong association with the development of obesity [395]. The family member Wnt5b also has been shown to have an elevated expression in adipose tissue, the pancreas, and the liver in patients with DM, suggesting control of metabolic pathways by Wnt [396]. Experimental animal studies with hyperglycemia following a high fat diet also show increased expression of Wnt3a and Wnt7a [397]. Intact Wnt family members may offer glucose tolerance and increased insulin sensitivity [398] as well as protect against DM complications such as renal glomerular mesangial cells injury [399]. In addition, animals that over express Wnt10b with a high-fat diet experienced a reduction in bodyweight, hyperinsulinemia, triglyceride plasma levels, and improved glucose homeostasis [400]. In contrast, patients with impaired Wnt signaling through a missense mutation in LRP-6 can have associated conditions with coronary artery disease and the combined metabolic syndrome with hypertension, hyperlipidemia, and DM [401]. This potential protective capacity of Wnt appears to extend to EPO [54, 254, 313]. Wnt1 protein is necessary and sufficient to promote cellular protection during elevated glucose exposure [54]. Administration of exogenous Wnt1 protein can significantly prevent apoptotic vascular cell injury during elevated glucose exposure. EPO also maintains the expression of Wnt1 during elevated glucose exposure and prevents loss of Wnt1 expression that would occur in the absence of EPO during elevated glucose. Blockade of Wnt1 with a Wnt1Ab can neutralize the protective capacity of EPO, demonstrating that Wnt1 is a critical component in the protective capacity of EPO during elevated glucose exposure [54].

FUTURE CONSIDERATIONS

Innovative investigations into the pathways that determine the dysfunction and injury of cellular systems during DM can lead the way for the development of new therapeutic inroads. In particular, both nicotinamide and EPO in conjunction with the signal transductions that are controlled by these agents offer novel platforms for drug discovery. Yet, these agents and their associated pathways also function in a broader sense as biomarkers for disease onset and progression. Biomarkers can involve the use of specific genes, proteins, or products of cellular and biological processes and may represent the response of cells or tissues to therapeutic strategies [62]. For example, loss of FoxO tissue expression may be a gauge for tumor progression [402] while enhanced FoxO protein expression may suggest an effective response to chemotherapy [361, 403, 404]. To a similar degree, the cellular expression of EPO and its receptor during injury paradigms also may indicate an effective treatment response [264, 290, 313], but during neoplastic growth may alternatively suggest a poor prognosis [405]. In regards to Wnt, the presence of this protein may be indicative of early tissue injury during elevated glucose [54], amyloid toxicity [32], or cardiac ischemia [406, 407]. Yet, the aberrant expression of Wnt under different circumstances may signal progressive prostate cancer and bone metastases [408], the induction of cancer stem cells [380], or the vulnerability of patients with Type 2 DM to develop colorectal tumors [409]. Future investigations into these pathways and the complexities that they hold will continue to channel clinical care into effective and innocuous strategies for both the prevention and treatment of DM.

Acknowledgments

This research was supported by the following grants to Kenneth Maiese: American Diabetes Association, American Heart Association (National), Bugher Foundation Award, Janssen Neuroscience Award, LEARN Foundation Award, MI Life Sciences Challenge Award, Nelson Foundation Award, NIH NIEHS (P30 ES06639), NIH NIA, NIH NINDS, and NIH ARRA.

References

  • 1.Maiese K, Chong ZZ, Shang YC. Mechanistic insights into diabetes mellitus and oxidative stress. Curr Med Chem. 2007;14(16):1729–38. doi: 10.2174/092986707781058968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Maiese K, Morhan SD, Chong ZZ. Oxidative stress biology and cell injury during type 1 and type 2 diabetes mellitus. Curr Neurovasc Res. 2007;4(1):63–71. doi: 10.2174/156720207779940653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Chan JC, Malik V, Jia W, et al. Diabetes in Asia: epidemiology, risk factors, and pathophysiology. JAMA. 2009;301(20):2129–40. doi: 10.1001/jama.2009.726. [DOI] [PubMed] [Google Scholar]
  • 4.Rebecchi KR, Wenke JL, Go EP, Desaire H. Label-free quantitation: a new glycoproteomics approach. J Am Soc Mass Spectrom. 2009;20(6):1048–59. doi: 10.1016/j.jasms.2009.01.013. [DOI] [PubMed] [Google Scholar]
  • 5.Maiese K. Diabetic stress: new triumphs and challenges to maintain vascular longevity. Expert Rev Cardiovasc Ther. 2008;6(3):281–4. doi: 10.1586/14779072.6.3.281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Daneman D. Type 1 diabetes. Lancet. 2006;367(9513):847–58. doi: 10.1016/S0140-6736(06)68341-4. [DOI] [PubMed] [Google Scholar]
  • 7.Dabelea D, Bell RA, D’Agostino RB, Jr, et al. Incidence of diabetes in youth in the United States. JAMA. 2007;297(24):2716–24. doi: 10.1001/jama.297.24.2716. [DOI] [PubMed] [Google Scholar]
  • 8.Development OfEC-oa. OECD Health Data; 2009. www.ecosante.org/oecd.htm. [Google Scholar]
  • 9.Hao J, Shen W, Tian C, et al. Mitochondrial nutrients improve immune dysfunction in the type 2 diabetic Goto-Kakizaki rats. J Cell Mol Med. 2009;13(4):701–11. doi: 10.1111/j.1582-4934.2008.00342.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Morley JE. Diabetes, sarcopenia, and frailty. Clin Geriatr Med. 2008;24(3):455–69. vi. doi: 10.1016/j.cger.2008.03.004. [DOI] [PubMed] [Google Scholar]
  • 11.McIntyre RS, Rasgon NL, Kemp DE, et al. Metabolic syndrome and major depressive disorder: co-occurrence and pathophysiologic overlap. Curr Diab Rep. 2009;9(1):51–9. doi: 10.1007/s11892-009-0010-0. [DOI] [PubMed] [Google Scholar]
  • 12.Wu SY, Wang GF, Liu ZQ, et al. Effect of geniposide, a hypoglycemic glucoside, on hepatic regulating enzymes in diabetic mice induced by a high-fat diet and streptozotocin. Acta Pharmacol Sin. 2009;30(2):202–8. doi: 10.1038/aps.2008.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Guarnieri G, Zanetti M, Vinci P, Cattin MR, Barazzoni R. Insulin resistance in chronic uremia. J Ren Nutr. 2009;19(1):20–4. doi: 10.1053/j.jrn.2008.11.014. [DOI] [PubMed] [Google Scholar]
  • 14.Aso Y, Suganuma R, Wakabayashi S, et al. Anemia is associated with an elevated serum level of high-molecular-weight adiponectin in patients with type 2 diabetes independently of renal dysfunction. Transl Res. 2009;154(4):175–82. doi: 10.1016/j.trsl.2009.07.005. [DOI] [PubMed] [Google Scholar]
  • 15.Gossai D, Lau-Cam CA. The effects of taurine, taurine homologs and hypotaurine on cell and membrane antioxidative system alterations caused by type 2 diabetes in rat erythrocytes. Adv Exp Med Biol. 2009;643:359–68. doi: 10.1007/978-0-387-75681-3_37. [DOI] [PubMed] [Google Scholar]
  • 16.Singh DK, Winocour P, Farrington K. Erythropoietic stress and anemia in diabetes mellitus. Nat Rev Endocrinol. 2009;5(4):204–10. doi: 10.1038/nrendo.2009.17. [DOI] [PubMed] [Google Scholar]
  • 17.Maiese K. Triple play: promoting neurovascular longevity with nicotinamide, WNT, and erythropoietin in diabetes mellitus. Biomed Pharmacother. 200;62(4):218–32. doi: 10.1016/j.biopha.2008.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Maiese K, Chong Z, Li F. Reducing oxidative stress and enhancing neurovascular longevity during diabetes mellitus. In: Maiese K, editor. Neurovascular Medicine: pursuing cellular longevity for healthy aging. New York NY: Oxford University Press; 2009. pp. 540–64. [Google Scholar]
  • 19.Donahoe SM, Stewart GC, McCabe CH, et al. Diabetes and mortality following acute coronary syndromes. JAMA. 2007;298(7):765–75. doi: 10.1001/jama.298.7.765. [DOI] [PubMed] [Google Scholar]
  • 20.Kuhad A, Bishnoi M, Tiwari V, Chopra K. Suppression of NF-kappabeta signaling pathway by tocotrienol can prevent diabetes associated cognitive deficits. Pharmacol Biochem Behav. 2009;92(2):251–9. doi: 10.1016/j.pbb.2008.12.012. [DOI] [PubMed] [Google Scholar]
  • 21.Maiese K, Chong ZZ, Hou J, Shang YC. The vitamin nicotinamide: translating nutrition into clinical care. Molecules. 2009;14(9):3446–85. doi: 10.3390/molecules14093446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Maiese K, Chong ZZ, Hou J, Shang YC. New strategies for Alzheimer’s disease and cognitive impairment. Oxid Med Cell Longev. 2009;2(5):279–90. doi: 10.4161/oxim.2.5.9990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Chong ZZ, Li F, Maiese K. Oxidative stress in the brain: Novel cellular targets that govern survival during neurodegenerative disease. Prog Neurobiol. 2005;75(3):207–46. doi: 10.1016/j.pneurobio.2005.02.004. [DOI] [PubMed] [Google Scholar]
  • 24.Chong ZZ, Li F, Maiese K. Stress in the brain: novel cellular mechanisms of injury linked to Alzheimer’s disease. Brain Res Brain Res Rev. 2005;49(1):1–21. doi: 10.1016/j.brainresrev.2004.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Maiese K, Holloway HH, Larson DM, Soncrant TT. Effect of acute and chronic arecoline treatment on cerebral metabolism and blood flow in the conscious rat. Brain Res. 1994;641(1):65–75. doi: 10.1016/0006-8993(94)91816-3. [DOI] [PubMed] [Google Scholar]
  • 26.Kim HS, Lim JY, Sul D, et al. Neuroprotective effects of the new diterpene, CBNU06 against beta-amyloid-induced toxicity through the inhibition of NF-kappaB signaling pathway in PC12 cells. Eur J Pharmacol. 2009;622(1–3):25–31. doi: 10.1016/j.ejphar.2009.09.007. [DOI] [PubMed] [Google Scholar]
  • 27.Salminen A, Kaarniranta K. Siglec receptors and hiding plaques in Alzheimer’s disease. J Mol Med. 2009;87(7):697–701. doi: 10.1007/s00109-009-0472-1. [DOI] [PubMed] [Google Scholar]
  • 28.Chong ZZ, Li F, Maiese K. Attempted Cell cycle induction in post-mitotic neurons occurs in early and late apoptotic programs through Rb, E2F1, and caspase 3. Curr Neurovasc Res. 2006;3(1):25–39. doi: 10.2174/156720206775541741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lin SH, Chong ZZ, Maiese K. Cell cycle induction in post-mitotic neurons proceeds in concert with the initial phase of programmed cell death in rat. Neurosci Lett. 2001;310(2–3):173–7. doi: 10.1016/s0304-3940(01)02118-8. [DOI] [PubMed] [Google Scholar]
  • 30.Majd S, Zarifkar A, Rastegar K, Takhshid MA. Different fibrillar Abeta 1–42 concentrations induce adult hippocampal neurons to reenter various phases of the cell cycle. Brain Res. 2008;1218:224–9. doi: 10.1016/j.brainres.2008.04.050. [DOI] [PubMed] [Google Scholar]
  • 31.Chong ZZ, Li F, Maiese K. Erythropoietin requires NF-kappaB and its nuclear translocation to prevent early and late apoptotic neuronal injury during beta-amyloid toxicity. Curr Neurovasc Res. 2005;2(5):387–99. doi: 10.2174/156720205774962683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Chong ZZ, Li F, Maiese K. Cellular demise and inflammatory microglial activation during beta-amyloid toxicity are governed by Wnt1 and canonical signaling pathways. Cell Signal. 2007;19(6):1150–62. doi: 10.1016/j.cellsig.2006.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Majd S, Rastegar K, Zarifkar A, Takhshid MA. Fibrillar β-amyloid (Aβ) (1–42) elevates extracellular Aβ in cultured hippocampal neurons of adult rats. Brain Res. 2007;1185:321–7. doi: 10.1016/j.brainres.2007.09.033. [DOI] [PubMed] [Google Scholar]
  • 34.Shang YC, Chong ZZ, Hou J, Maiese K. The forkhead transcription factor FoxO3a controls microglial inflammatory activation and eventual apoptotic injury through caspase 3. Curr Neurovasc Res. 2009;6(1):20–31. doi: 10.2174/156720209787466064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Bakshi P, Margenthaler E, Laporte V, Crawford F, Mullan M. Novel role of CXCR2 in regulation of gamma-secretase activity. ACS Chem Biol. 2008;3(12):777–89. doi: 10.1021/cb800167a. [DOI] [PubMed] [Google Scholar]
  • 36.Lu J, Wu DM, Zheng YL, et al. Trace amounts of copper exacerbate beta amyloid-induced neurotoxicity in the cholesterol-fed mice through TNF-mediated inflammatory pathway. Brain Behav Immun. 2009;23(2):193–203. doi: 10.1016/j.bbi.2008.09.003. [DOI] [PubMed] [Google Scholar]
  • 37.Bitner RS, Nikkel AL, Markosyan S, et al. Selective alpha7 nicotinic acetylcholine receptor activation regulates glycogen synthase kinase3β and decreases tau phosphorylation in vivo. Brain Res. 2009;1265:65–74. doi: 10.1016/j.brainres.2009.01.069. [DOI] [PubMed] [Google Scholar]
  • 38.Vaisid T, Barnoy S, Kosower NS. Calpastatin overexpression attenuates amyloid-β-peptide toxicity in differentiated PC12 cells. Neuroscience. 2008;156(4):921–31. doi: 10.1016/j.neuroscience.2008.07.072. [DOI] [PubMed] [Google Scholar]
  • 39.Erol A. Unraveling the molecular mechanisms behind the metabolic basis of sporadic Alzheimer’s disease. J Alzheimers Dis. 2009;17(2):267–76. doi: 10.3233/JAD-2009-1047. [DOI] [PubMed] [Google Scholar]
  • 40.Maiese K, Chong ZZ. Nicotinamide: necessary nutrient emerges as a novel cytoprotectant for the brain. Trends Pharmacol Sci. 2003;24(5):228–32. doi: 10.1016/S0165-6147(03)00078-6. [DOI] [PubMed] [Google Scholar]
  • 41.Schubert M, Gautam D, Surjo D, et al. Role for neuronal insulin resistance in neurodegenerative diseases. Proc Natl Acad Sci USA. 2004;101(9):3100–5. doi: 10.1073/pnas.0308724101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Beeri MS, Silverman JM, Davis KL, et al. Type 2 diabetes is negatively associated with Alzheimer’s disease neuropathology. J Gerontol A Biol Sci Med Sci. 2005;60(4):471–5. doi: 10.1093/gerona/60.4.471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Luchsinger JA, Tang MX, Stern Y, Shea S, Mayeux R. Diabetes mellitus and risk of Alzheimer’s disease and dementia with stroke in a multiethnic cohort. Am J Epidemiol. 2001;154(7):635–41. doi: 10.1093/aje/154.7.635. [DOI] [PubMed] [Google Scholar]
  • 44.Steen E, Terry BM, Rivera EJ, et al. Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer’s disease--is this type 3 diabetes? J Alzheimers Dis. 2005;7(1):63–80. doi: 10.3233/jad-2005-7107. [DOI] [PubMed] [Google Scholar]
  • 45.Newsholme P, Haber EP, Hirabara SM, et al. Diabetes associated cell stress and dysfunction: role of mitochondrial and non-mitochondrial ROS production and activity. J Physiol. 2007;583(Pt 1):9–24. doi: 10.1113/jphysiol.2007.135871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Szabo C. Role of nitrosative stress in the pathogenesis of diabetic vascular dysfunction. Br J Pharmacol. 2009;156(5):713–27. doi: 10.1111/j.1476-5381.2008.00086.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Simone S, Gorin Y, Velagapudi C, Abboud HE, Habib SL. Mechanism of oxidative DNA damage in diabetes: tuberin inactivation and downregulation of DNA repair enzyme 8-oxo-7,8-dihydro-2′-deoxyguanosine-DNA glycosylase. Diabetes. 2008;57(10):2626–36. doi: 10.2337/db07-1579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Gaddini L, Villa M, Matteucci A, et al. Early effects of high glucose in retinal tissue cultures Renin-Angiotensin system-dependent and -independent signaling. Neurobiol Dis. 2009;35(2):278–85. doi: 10.1016/j.nbd.2009.05.013. [DOI] [PubMed] [Google Scholar]
  • 49.Singh DK, Winocour P, Farrington K. Mechanisms of disease: the hypoxic tubular hypothesis of diabetic nephropathy. Nat Clin Pract Nephrol. 2008;4(4):216–26. doi: 10.1038/ncpneph0757. [DOI] [PubMed] [Google Scholar]
  • 50.Lin J, Zheng S, Chen A. Curcumin attenuates the effects of insulin on stimulating hepatic stellate cell activation by interrupting insulin signaling and attenuating oxidative stress. Lab Invest. 2009;89(12):1397–409. doi: 10.1038/labinvest.2009.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Barbosa NB, Oliveira C, Araldi D, et al. Acute diphenyl diselenide treatment reduces hyperglycemia but does not change delta-aminolevulinate dehydratase activity in alloxan-induced diabetes in rats. Biol Pharm Bull. 2008;31(12):2200–4. doi: 10.1248/bpb.31.2200. [DOI] [PubMed] [Google Scholar]
  • 52.Memisogullari R, Bakan E. Levels of ceruloplasmin, transferrin, and lipid peroxidation in the serum of patients with type 2 diabetes mellitus. J Diabetes Comp. 2004;18(4):193–7. doi: 10.1016/S1056-8727(03)00032-1. [DOI] [PubMed] [Google Scholar]
  • 53.Chattopadhyay M, Walter C, Mata M, Fink DJ. Neuroprotective effect of herpes simplex virus-mediated gene transfer of erythropoietin in hyperglycemic dorsal root ganglion neurons. Brain. 2009;132(Pt 4):879–88. doi: 10.1093/brain/awp014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Chong ZZ, Shang YC, Maiese K. Vascular injury during elevated glucose can be mitigated by erythropoietin and Wnt signaling. Curr Neurovasc Res. 2007;4(3):194–204. doi: 10.2174/156720207781387150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Liu W, Liu P, Tao S, et al. Berberine inhibits aldose reductase and oxidative stress in rat mesangial cells cultured under high glucose. Arch Biochem Biophys. 2008;475(2):128–34. doi: 10.1016/j.abb.2008.04.022. [DOI] [PubMed] [Google Scholar]
  • 56.Shang YC, Chong ZZ, Hou J, Maiese K. FoxO3a governs early microglial proliferation and employs mitochondrial depolarization with caspase 3, 8, and 9 cleavage during oxidant induced apoptosis. Curr Neurovasc Res. 2009;6(4):223–38. doi: 10.2174/156720209789630302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Chen H, Li X, Epstein PN. MnSOD and catalase transgenes demonstrate that protection of islets from oxidative stress does not alter cytokine toxicity. Diabetes. 2005;54(5):1437–46. doi: 10.2337/diabetes.54.5.1437. [DOI] [PubMed] [Google Scholar]
  • 58.Lepore DA, Shinkel TA, Fisicaro N, et al. Enhanced expression of glutathione peroxidase protects islet beta cells from hypoxia-reoxygenation. Xenotransplantation. 2004;11(1):53–9. doi: 10.1111/j.1399-3089.2004.00082.x. [DOI] [PubMed] [Google Scholar]
  • 59.Ribeiro MC, Barbosa NB, de Almeida TM, et al. High-fat diet and hydrochlorothiazide increase oxidative stress in brain of rats. Cell Biochem Funct. 2009;27(7):473–8. doi: 10.1002/cbf.1599. [DOI] [PubMed] [Google Scholar]
  • 60.Rachek LI, Thornley NP, Grishko VI, LeDoux SP, Wilson GL. Protection of INS-1 cells from free fatty acid-induced apoptosis by targeting hOGG1 to mitochondria. Diabetes. 2006;55(4):1022–8. doi: 10.2337/diabetes.55.04.06.db05-0865. [DOI] [PubMed] [Google Scholar]
  • 61.Haber CA, Lam TK, Yu Z, et al. N-acetylcysteine and taurine prevent hyperglycemia-induced insulin resistance in vivo: possible role of oxidative stress. Am J Physiol Endocrinol Metab. 2003;285(4):E744–53. doi: 10.1152/ajpendo.00355.2002. [DOI] [PubMed] [Google Scholar]
  • 62.Maiese K. Marking the onset of oxidative stress: Biomarkers and novel strategies. Oxid Med Cell Longev. 2009;2(1):1. doi: 10.4161/oxim.2.1.8059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Maiese K, Hou J, Chong ZZ, Shang YC. Erythropoietin, forkhead proteins, and oxidative injury: biomarkers and biology. Sci World J. 2009;9:1072–104. doi: 10.1100/tsw.2009.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Maiese K, Chong ZZ, Hou J, Shang YC. Erythropoietin and oxidative stress. Curr Neurovasc Res. 2008;5(2):125–42. doi: 10.2174/156720208784310231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Yano M, Hasegawa G, Ishii M, et al. Short-term exposure of high glucose concentration induces generation of reactive oxygen species in endothelial cells: implication for the oxidative stress associated with postprandial hyperglycemia. Redox Rep. 2004;9(2):111–6. doi: 10.1179/135100004225004779. [DOI] [PubMed] [Google Scholar]
  • 66.Monnier L, Mas E, Ginet C, et al. Activation of oxidative stress by acute glucose fluctuations compared with sustained chronic hyperglycemia in patients with type 2 diabetes. JAMA. 2006;295(14):1681–7. doi: 10.1001/jama.295.14.1681. [DOI] [PubMed] [Google Scholar]
  • 67.De Felice FG, Velasco PT, Lambert MP, et al. Aβ oligomers induce neuronal oxidative stress through an N-methyl-D-aspartate receptor-dependent mechanism that is blocked by the Alzheimer drug memantine. J Biol Chem. 2007;282(15):11590–601. doi: 10.1074/jbc.M607483200. [DOI] [PubMed] [Google Scholar]
  • 68.Lin SH, Maiese K. The metabotropic glutamate receptor system protects against ischemic free radical programmed cell death in rat brain endothelial cells. J Cereb Blood Flow Metab. 2001;21(3):262–75. doi: 10.1097/00004647-200103000-00010. [DOI] [PubMed] [Google Scholar]
  • 69.Chuang HH, Lin S. Oxidative challenges sensitize the capsaicin receptor by covalent cysteine modification. Proc Natl Acad Sci USA. 2009;106(47):20097–102. doi: 10.1073/pnas.0902675106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Nomoto M, Miyata M, Yin S, et al. Bile acid-induced elevated oxidative stress in the absence of farnesoid X receptor. Biol Pharm Bull. 2009;32(2):172–8. doi: 10.1248/bpb.32.172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Walsh KB, Toledo AH, Rivera-Chavez FA, Lopez-Neblina F, Toledo-Pereyra LH. Inflammatory mediators of liver ischemia-reperfusion injury. Exp Clin Transplant. 2009;7(2):78–93. [PubMed] [Google Scholar]
  • 72.Escobar J, Pereda J, Arduini A, et al. Cross-talk between oxidative stress and pro-inflammatory cytokines in acute pancreatitis: a key role for protein phosphatases. Curr Pharm Des. 2009;15(26):3027–42. doi: 10.2174/138161209789058075. [DOI] [PubMed] [Google Scholar]
  • 73.Bouayed J, Rammal H, Soulimani R. Oxidative stress and anxiety: Relationship and cellular pathways. Oxid Med Cell Longev. 2009;2(2):63–7. doi: 10.4161/oxim.2.2.7944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Maiese K. High anxiety: recognizing stress as the stressor. Oxid Med Cell Longev. 2009;2(2):61–2. doi: 10.4161/oxim.2.2.8842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Campos-Esparza MR, Sanchez-Gomez MV, Matute C. Molecular mechanisms of neuroprotection by two natural antioxidant polyphenols. Cell Calcium. 2009;45(4):358–68. doi: 10.1016/j.ceca.2008.12.007. [DOI] [PubMed] [Google Scholar]
  • 76.Chong ZZ, Li F, Maiese K. Group I metabotropic receptor neuroprotection requires Akt and its substrates that govern FOXO3a, Bim, and β-catenin during oxidative stress. Curr Neurovasc Res. 2006;3(2):107–17. doi: 10.2174/156720206776875830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.He Z, Lu Q, Xu X, et al. DDPH ameliorated oxygen and glucose deprivation-induced injury in rat hippocampal neurons via interrupting Ca2+ overload and glutamate release. Eur J Pharmacol. 2009;603(1–3):50–5. doi: 10.1016/j.ejphar.2008.12.010. [DOI] [PubMed] [Google Scholar]
  • 78.Lehtinen MK, Tegelberg S, Schipper H, et al. Cystatin B deficiency sensitizes neurons to oxidative stress in progressive myoclonus epilepsy, EPM1. J Neurosci. 2009;29(18):5910–5. doi: 10.1523/JNEUROSCI.0682-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Ye J, Han Y, Wang C, Yu W. Cytoprotective effect of polypeptide from Chlamys farreri on neuroblastoma (SH-SY5Y) cells following HO exposure involves scavenging ROS and inhibition JNK phosphorylation. J Neurochem. 2009;111(2):441–51. doi: 10.1111/j.1471-4159.2009.06328.x. [DOI] [PubMed] [Google Scholar]
  • 80.Anderson DW, Bradbury KA, Schneider JS. Broad neuroprotective profile of nicotinamide in different mouse models of MPTP-induced parkinsonism. Eur J Neurosci. 2008;28(3):610–7. doi: 10.1111/j.1460-9568.2008.06356.x. [DOI] [PubMed] [Google Scholar]
  • 81.Morissette M, Al Sweidi S, Callier S, Di Paolo T. Estrogen and SERM neuroprotection in animal models of Parkinson’s disease. Mol Cell Endocrinol. 2008;290(1–2):60–9. doi: 10.1016/j.mce.2008.04.008. [DOI] [PubMed] [Google Scholar]
  • 82.Morissette M, Le Saux M, D’Astous M, et al. Contribution of estrogen receptors alpha and beta to the effects of estradiol in the brain. J Steroid Biochem Mol Biol. 2008;108(3–5):327–38. doi: 10.1016/j.jsbmb.2007.09.011. [DOI] [PubMed] [Google Scholar]
  • 83.Rodriguez-Blanco J, Martin V, Herrera F, et al. Intracellular signaling pathways involved in post-mitotic dopaminergic PC12 cell death induced by 6-hydroxydopamine. J Neurochem. 2008;107(1):127–40. doi: 10.1111/j.1471-4159.2008.05588.x. [DOI] [PubMed] [Google Scholar]
  • 84.Sales Santos I, da Rocha Tomé A, Saldanha G, et al. Oxidative stress in the hippocampus during experimental seizures can be ameliorated with the antioxidant ascorbic acid. Oxid Med Cell Longev. 2009;2(4):23–30. doi: 10.4161/oxim.2.4.8876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Maiese K, Chong ZZ, Shang YC, Hou J. Therapeutic promise and principles: Metabotropic glutamate receptors. Oxid Med Cell Longev. 2008;1(1):1–14. doi: 10.4161/oxim.1.1.6842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Probst-Hensch NM, Imboden M, Felber Dietrich D, et al. Glutathione S-transferase polymorphisms, passive smoking, obesity, and heart rate variability in nonsmokers. Environ Health Perspect. 2008;116(11):1494–9. doi: 10.1289/ehp.11402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Yemisci M, Gursoy-Ozdemir Y, Vural A, et al. Pericyte contraction induced by oxidative-nitrative stress impairs capillary reflow despite successful opening of an occluded cerebral artery. Nat Med. 2009;15(9):1031–7. doi: 10.1038/nm.2022. [DOI] [PubMed] [Google Scholar]
  • 88.Fatma N, Kubo E, Toris CB, et al. PRDX6 attenuates oxidative stress- and TGFbeta-induced abnormalities of human trabecular meshwork cells. Free Radic Res. 2009;43(9):783–95. doi: 10.1080/10715760903062887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Gomes P, Simão S, Silva E, et al. Aging increases oxidative stress and renal expression of oxidant and antioxidant enzymes that are associated with an increased trend in systolic blood pressure. Oxid Med Cell Longev. 2009;2(3):19–26. doi: 10.4161/oxim.2.3.8819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Pedersen MO, Jensen R, Pedersen DS, et al. Metallothionein-I+II in neuroprotection. BioFactors (Oxford, England) 2009;35(4):315–25. doi: 10.1002/biof.44. [DOI] [PubMed] [Google Scholar]
  • 91.Astiz M, de Alaniz MJ, Marra CA. Effect of pesticides on cell survival in liver and brain rat tissues. Ecotoxicol Environ Saf. 2009;72(7):2025–32. doi: 10.1016/j.ecoenv.2009.05.001. [DOI] [PubMed] [Google Scholar]
  • 92.Slotkin TA, Seidler FJ. Protein kinase C is a target for diverse developmental neurotoxicants: transcriptional responses to chlorpyrifos, diazinon, dieldrin and divalent nickel in PC12 cells. Brain Res. 2009;1263:23–32. doi: 10.1016/j.brainres.2009.01.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Hamden K, Carreau S, Jamoussi K, et al. 1α,25 dihydroxyvitamin D3: therapeutic and preventive effects against oxidative stress, hepatic, pancreatic and renal injury in alloxan-induced diabetes in rats. J Nutr Sci Vitaminol (Tokyo) 2009;55(3):215–22. doi: 10.3177/jnsv.55.215. [DOI] [PubMed] [Google Scholar]
  • 94.Regulska M, Leskiewicz M, Budziszewska B, et al. Inhibitory effects of 1,25-dihydroxyvitamin D(3) and its low-calcemic analogues on staurosporine-induced apoptosis. Pharmacol Rep. 2007;59(4):393–401. [PubMed] [Google Scholar]
  • 95.Li F, Chong ZZ, Maiese K. Cell life versus cell longevity: the mysteries surrounding the NAD(+) precursor nicotinamide. Curr Med Chem. 2006;13(8):883–95. doi: 10.2174/092986706776361058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Li J, Wang H, Rosenberg PA. Vitamin K prevents oxidative cell death by inhibiting activation of 12-lipoxygenase in developing oligodendrocytes. J Neurosci Res. 2009;87(9):1997–2005. doi: 10.1002/jnr.22029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Ozsoy N, Candoken E, Akev N. Implications for degenerative disorders: Antioxidative activity, total phenols, flavonoids, ascorbic acid, β-carotene, α-tocopherol in Aloe vera. Oxid Med Cell Longev. 2009;2(2):99–106. doi: 10.4161/oxim.2.2.8493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Then SM, Mazlan M, Mat Top G, Wan Ngah WZ. Is vitamin E toxic to neuron cells? Cell Mol Neurobiol. 2009;29(4):485–96. doi: 10.1007/s10571-008-9340-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.El-Mir MY, Detaille D, GRV, et al. Neuroprotective role of antidiabetic drug metformin against apoptotic cell death in primary cortical neurons. J Mol Neurosci. 2008;34(1):77–87. doi: 10.1007/s12031-007-9002-1. [DOI] [PubMed] [Google Scholar]
  • 100.Kui L, Weiwei Z, Ling L, et al. Ghrelin inhibits apoptosis induced by high glucose and sodium palmitate in adult rat cardiomyocytes through the PI3K-Akt signaling pathway. Regul Pept. 2009;155(1–3):62–9. doi: 10.1016/j.regpep.2009.03.003. [DOI] [PubMed] [Google Scholar]
  • 101.Head BP, Patel HH, Niesman IR, et al. Inhibition of p75 neurotrophin receptor attenuates isoflurane-mediated neuronal apoptosis in the neonatal central nervous system. Anesthesiology. 2009;110(4):813–25. doi: 10.1097/ALN.0b013e31819b602b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Han Z, Xiao MJ, Shao B, et al. Attenuation of ischemia-induced rat brain injury by 2-(-2-benzofuranyl)-2-imidazoline, a high selectivity ligand for imidazoline I(2) receptors. Neurol Res. 2009;31(4):390–5. doi: 10.1179/174313209X444116. [DOI] [PubMed] [Google Scholar]
  • 103.Maiese K. From the bench to the bedside: the molecular management of cerebral ischemia. Clin Neuropharm. 1998;21(1):1–7. [PubMed] [Google Scholar]
  • 104.Maiese K, Pek L, Berger SB, Reis DJ. Reduction in focal cerebral ischemia by agents acting at imidazole receptors. J Cereb Blood Flow Metab. 1992;12(1):53–63. doi: 10.1038/jcbfm.1992.7. [DOI] [PubMed] [Google Scholar]
  • 105.Nakka VP, Gusain A, Mehta SL, Raghubir R. Molecular mechanisms of apoptosis in cerebral ischemia: multiple neuroprotective opportunities. Mol Neurobiol. 2008;37(1):7–38. doi: 10.1007/s12035-007-8013-9. [DOI] [PubMed] [Google Scholar]
  • 106.Cardoso L, Herman BC, Verborgt O, et al. Osteocyte apoptosis controls activation of intracortical resorption in response to bone fatigue. J Bone Miner Res. 2009;24(4):597–605. doi: 10.1359/JBMR.081210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Arboleda G, Morales LC, Benitez B, Arboleda H. Regulation of ceramide-induced neuronal death: cell metabolism meets neurodegeneration. Brain Res Rev. 2009;59(2):333–46. doi: 10.1016/j.brainresrev.2008.10.001. [DOI] [PubMed] [Google Scholar]
  • 108.Maiese K, Vincent AM. Critical temporal modulation of neuronal programmed cell injury. Cell Mol Neurobiol. 2000;20(3):383–400. doi: 10.1023/A:1007070311203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Zhong J, Zheng W, Huang L, et al. PrP106–126 amide causes the semi-penetrated poration in the supported lipid bilayers. Biochim Biophys Acta. 2007;1768(6):1420–9. doi: 10.1016/j.bbamem.2007.03.003. [DOI] [PubMed] [Google Scholar]
  • 110.Burgos-Ramos E, Puebla-Jimenez L, Arilla-Ferreiro E. Minocycline provides protection against β-amyloid(25–35)-induced alterations of the somatostatin signaling pathway in the rat temporal cortex. Neuroscience. 2008;154(4):1458–66. doi: 10.1016/j.neuroscience.2008.04.036. [DOI] [PubMed] [Google Scholar]
  • 111.Burgos-Ramos E, Puebla-Jimenez L, Arilla-Ferreiro E. Minocycline prevents Aβ(25–35)-induced reduction of somatostatin and neprilysin content in rat temporal cortex. Life Sci. 2009;84(7–8):205–10. doi: 10.1016/j.lfs.2008.11.019. [DOI] [PubMed] [Google Scholar]
  • 112.Casoli T, Di Stefano G, Giorgetti B, et al. Release of β-amyloid from high-density platelets: implications for Alzheimer’s disease pathology. Ann NY Acad Sci. 2007;1096:170–8. doi: 10.1196/annals.1397.082. [DOI] [PubMed] [Google Scholar]
  • 113.Kelley BJ, Knopman DS. Alternative medicine and Alzheimer disease. Neurologist. 2008;14(5):299–306. doi: 10.1097/NRL.0b013e318172cf4d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Liang WS, Dunckley T, Beach TG, et al. Altered neuronal gene expression in brain regions differentially affected by Alzheimer’s disease: a reference data set. Physiol Genom. 2008;33(2):240–56. doi: 10.1152/physiolgenomics.00242.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Verdaguer E, Susana Gde A, Clemens A, Pallas M, Camins A. Implication of the transcription factor E2F-1 in the modulation of neuronal apoptosis. Biomed Pharmacother. 2007;61(7):390–9. doi: 10.1016/j.biopha.2006.11.001. [DOI] [PubMed] [Google Scholar]
  • 116.Sommer C. Neuronal plasticity after ischemic preconditioning and TIA-like preconditioning ischemic periods. Acta Neuropathol. 2009;117(5):511–23. doi: 10.1007/s00401-008-0473-0. [DOI] [PubMed] [Google Scholar]
  • 117.Braga M, Sinha Hikim AP, Datta S, et al. Involvement of oxidative stress and caspase 2-mediated intrinsic pathway signaling in age-related increase in muscle cell apoptosis in mice. Apoptosis. 2008;13(6):822–32. doi: 10.1007/s10495-008-0216-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Maiese K, Chong ZZ, Kang J. Transformation into treatment: novel therapeutics that begin within the cell. In: Maiese K, editor. Neuronal and vascular plasticity: elucidating basic cellular mechanisms for future therapeutic discovery. Norwell, MA: Kluwer Academic Publishers; 2003. pp. 1–26. [Google Scholar]
  • 119.Gross J, Machulik A, Amarjargal N, et al. Expression of apoptosis-related genes in the organ of Corti, modiolus and stria vascularis of newborn rats. Brain Res. 2007;1162:56–68. doi: 10.1016/j.brainres.2007.05.061. [DOI] [PubMed] [Google Scholar]
  • 120.Maiese K, Ahmad I, TenBroeke M, Gallant J. Metabotropic glutamate receptor subtypes independently modulate neuronal intracellular calcium. J Neurosci Res. 1999;55:472–85. doi: 10.1002/(SICI)1097-4547(19990215)55:4<472::AID-JNR7>3.0.CO;2-Q. [DOI] [PubMed] [Google Scholar]
  • 121.Maiese K, Vincent AM. Membrane asymmetry and DNA degradation: functionally distinct determinants of neuronal programmed cell death. J Neurosci Res. 2000;59(4):568–80. doi: 10.1002/(SICI)1097-4547(20000215)59:4<568::AID-JNR13>3.0.CO;2-R. [DOI] [PubMed] [Google Scholar]
  • 122.Chong ZZ, Kang JQ, Maiese K. Metabotropic glutamate receptors promote neuronal and vascular plasticity through novel intracellular pathways. Histol Histopathol. 2003;18(1):173–89. doi: 10.14670/HH-18.173. [DOI] [PubMed] [Google Scholar]
  • 123.Dombroski D, Balasubramanian K, Schroit AJ. Phosphatidylserine expression on cell surfaces promotes antibody- dependent aggregation and thrombosis in β2-glycoprotein I-immune mice. J Autoimmun. 2000;14(3):221–9. doi: 10.1006/jaut.2000.0365. [DOI] [PubMed] [Google Scholar]
  • 124.Jessel R, Haertel S, Socaciu C, Tykhonova S, Diehl HA. Kinetics of apoptotic markers in exogeneously induced apoptosis of EL4 cells. J Cell Mol Med. 2002;6(1):82–92. doi: 10.1111/j.1582-4934.2002.tb00313.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Kang JQ, Chong ZZ, Maiese K. Critical role for Akt1 in the modulation of apoptotic phosphatidylserine exposure and microglial activation. Mol Pharmacol. 2003;64(3):557–69. doi: 10.1124/mol.64.3.557. [DOI] [PubMed] [Google Scholar]
  • 126.Kang JQ, Chong ZZ, Maiese K. Akt1 protects against inflammatory microglial activation through maintenance of membrane asymmetry and modulation of cysteine protease activity. J Neurosci Res. 2003;74(1):37–51. doi: 10.1002/jnr.10740. [DOI] [PubMed] [Google Scholar]
  • 127.Mari C, Karabiyikoglu M, Goris ML, et al. Detection of focal hypoxic-ischemic injury and neuronal stress in a rodent model of unilateral MCA occlusion/reperfusion using radiolabeled annexin V. Eur J Nucl Med Mol Imaging. 2004;31(5):733–9. doi: 10.1007/s00259-004-1473-5. [DOI] [PubMed] [Google Scholar]
  • 128.Maiese K. The dynamics of cellular injury: transformation into neuronal and vascular protection. Histol Histopathol. 2001;16(2):633–44. doi: 10.14670/HH-16.633. [DOI] [PubMed] [Google Scholar]
  • 129.Vincent AM, Maiese K. Direct temporal analysis of apoptosis induction in living adherent neurons. J Histochem Cytochem. 1999;47(5):661–72. doi: 10.1177/002215549904700508. [DOI] [PubMed] [Google Scholar]
  • 130.Chong ZZ, Lin SH, Kang JQ, Maiese K. Erythropoietin prevents early and late neuronal demise through modulation of Akt1 and induction of caspase 1, 3, and 8. J Neurosci Res. 2003;71(5):659–69. doi: 10.1002/jnr.10528. [DOI] [PubMed] [Google Scholar]
  • 131.Chong ZZ, Lin SH, Kang JQ, Maiese K. The tyrosine phosphatase SHP2 modulates MAP kinase p38 and caspase 1 and 3 to foster neuronal survival. Cell Mol Neurobiol. 2003;23(4–5):561–78. doi: 10.1023/A:1025158314016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Maiese K, Boccone L. Neuroprotection by peptide growth factors against anoxia and nitric oxide toxicity requires modulation of protein kinase C. J Cereb Blood Flow Metab. 1995;15(3):440–9. doi: 10.1038/jcbfm.1995.55. [DOI] [PubMed] [Google Scholar]
  • 133.Maiese K, Boniece IR, Skurat K, Wagner JA. Protein kinases modulate the sensitivity of hippocampal neurons to nitric oxide toxicity and anoxia. J Neurosci Res. 1993;36(1):77–87. doi: 10.1002/jnr.490360109. [DOI] [PubMed] [Google Scholar]
  • 134.Maiese K, TenBroeke M, Kue I. Neuroprotection of lubeluzole is mediated through the signal transduction pathways of nitric oxide. J Neurochem. 1997;68(2):710–4. doi: 10.1046/j.1471-4159.1997.68020710.x. [DOI] [PubMed] [Google Scholar]
  • 135.Leytin V, Allen DJ, Mykhaylov S, Lyubimov E, Freedman J. Thrombin-triggered platelet apoptosis. J Thromb Haemost. 2006;4(12):2656–63. doi: 10.1111/j.1538-7836.2006.02200.x. [DOI] [PubMed] [Google Scholar]
  • 136.Gilfillan AM, Rivera J. The tyrosine kinase network regulating mast cell activation. Immunol Rev. 2009;228(1):149–69. doi: 10.1111/j.1600-065X.2008.00742.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Maiese K, Chong ZZ, Shang YC, Hou J. A “FOXO” in sight: targeting Foxo proteins from conception to cancer. Med Res Rev. 2009;29(3):395–418. doi: 10.1002/med.20139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Chong ZZ, Li F, Maiese K. The pro-survival pathways of mTOR and protein kinase B target glycogen synthase kinase-3β and nuclear factor-kappaB to foster endogenous microglial cell protection. Int J Mol Med. 2007;19(2):263–72. [PMC free article] [PubMed] [Google Scholar]
  • 139.Dello Russo C, Lisi L, Tringali G, Navarra P. Involvement of mTOR kinase in cytokine-dependent microglial activation and cell proliferation. Biochem Pharmacol. 2009;78(9):1242–51. doi: 10.1016/j.bcp.2009.06.097. [DOI] [PubMed] [Google Scholar]
  • 140.Lee SJ, Cho KS, Koh JY. Oxidative injury triggers autophagy in astrocytes: the role of endogenous zinc. Glia. 2009;57(12):1351–61. doi: 10.1002/glia.20854. [DOI] [PubMed] [Google Scholar]
  • 141.Martin I, Andres CR, Vedrine S, et al. Effect of the oligodendrocyte myelin glycoprotein (OMgp) on the expansion and neuronal differentiation of rat neural stem cells. Brain Res. 2009;1284:22–30. doi: 10.1016/j.brainres.2009.05.070. [DOI] [PubMed] [Google Scholar]
  • 142.Geijtenbeek TB, Gringhuis SI. Signalling through C-type lectin receptors: shaping immune responses. Nat Rev Immunol. 2009;9(7):465–79. doi: 10.1038/nri2569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Williams R, Dhillon NK, Hegde ST, et al. Proinflammatory cytokines and HIV-1 synergistically enhance CXCL10 expression in human astrocytes. Glia. 2009;57(7):734–43. doi: 10.1002/glia.20801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Zhao L, Ma W, Fariss RN, Wong WT. Retinal vascular repair and neovascularization are not dependent on CX3CR1 signaling in a model of ischemic retinopathy. Exp Eye Res. 2009;88(6):1004–13. doi: 10.1016/j.exer.2008.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Dringen R. Oxidative and antioxidative potential of brain microglial cells. Antioxid Redox Signal. 2005;7(9–10):1223–33. doi: 10.1089/ars.2005.7.1223. [DOI] [PubMed] [Google Scholar]
  • 146.Maiese K, Chong ZZ. Insights into oxidative stress and potential novel therapeutic targets for Alzheimer disease. Restor Neurol Neurosci. 2004;22(2):87–104. [PubMed] [Google Scholar]
  • 147.Sankarapandi S, Zweier JL, Mukherjee G, Quinn MT, Huso DL. Measurement and characterization of superoxide generation in microglial cells: evidence for an NADPH oxidase-dependent pathway. Arch Biochem Biophys. 1998;353(2):312–21. doi: 10.1006/abbi.1998.0658. [DOI] [PubMed] [Google Scholar]
  • 148.Denes A, Ferenczi S, Halasz J, Kornyei Z, Kovacs KJ. Role of CX3CR1 (fractalkine receptor) in brain damage and inflammation induced by focal cerebral ischemia in mouse. J Cereb Blood Flow Metab. 2008;28(10):1707–21. doi: 10.1038/jcbfm.2008.64. [DOI] [PubMed] [Google Scholar]
  • 149.Wu Y, Peng H, Cui M, et al. CXCL12 increases human neural progenitor cell proliferation through Akt-1/FOXO3a signaling pathway. J Neurochem. 2009;109(4):1157–67. doi: 10.1111/j.1471-4159.2009.06043.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Mallat M, Marin-Teva JL, Cheret C. Phagocytosis in the developing CNS: more than clearing the corpses. Curr Opin Neurobiol. 2005;15(1):101–7. doi: 10.1016/j.conb.2005.01.006. [DOI] [PubMed] [Google Scholar]
  • 151.Chong ZZ, Kang J, Li F, Maiese K. mGluRI targets microglial activation and selectively prevents neuronal cell engulfment through akt and caspase dependent pathways. Curr Neurovasc Res. 2005;2(3):197–211. doi: 10.2174/1567202054368317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Li F, Chong ZZ, Maiese K. Microglial integrity is maintained by erythropoietin through integration of Akt and its substrates of glycogen synthase kinase-3β, β-catenin, and nuclear factor-kappaB. Curr Neurovasc Res. 2006;3(3):187–201. doi: 10.2174/156720206778018758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Li F, Chong ZZ, Maiese K. Winding through the WNT pathway during cellular development and demise. Histol Histopathol. 2006;21(1):103–24. doi: 10.14670/hh-21.103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.He XL, Wang YH, Gao M, et al. Baicalein protects rat brain mitochondria against chronic cerebral hypoperfusion-induced oxidative damage. Brain Res. 2009;1249:212–21. doi: 10.1016/j.brainres.2008.10.005. [DOI] [PubMed] [Google Scholar]
  • 155.Maiese K, Chong ZZ, Shang YC, Hou J. Rogue proliferation versus restorative protection: where do we draw the line for Wnt and forkhead signaling? Expert opinion on therapeutic targets. 2008;12(7):905–16. doi: 10.1517/14728222.12.7.905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Plecita-Hlavata L, Lessard M, Santorova J, Bewersdorf J, Jezek P. Mitochondrial oxidative phosphorylation and energetic status are reflected by morphology of mitochondrial network in INS-1E and HEP-G2 cells viewed by 4Pi microscopy. Biochim Biophys Acta. 2008;1777(7–8):834–46. doi: 10.1016/j.bbabio.2008.04.002. [DOI] [PubMed] [Google Scholar]
  • 157.Di Lisa F, Menabo R, Canton M, Barile M, Bernardi P. Opening of the mitochondrial permeability transition pore causes depletion of mitochondrial and cytosolic NAD+ and is a causative event in the death of myocytes in postischemic reperfusion of the heart. J Biol Chem. 2001;276(4):2571–5. doi: 10.1074/jbc.M006825200. [DOI] [PubMed] [Google Scholar]
  • 158.Chong ZZ, Kang JQ, Maiese K. Apaf-1, Bcl-xL, Cytochrome c, and Caspase-9 form the critical elements for cerebral vascular protection by erythropoietin. J Cereb Blood Flow Metab. 2003;23(3):320–30. doi: 10.1097/01.WCB.0000050061.57184.AE. [DOI] [PubMed] [Google Scholar]
  • 159.Lin SH, Vincent A, Shaw T, Maynard KI, Maiese K. Prevention of nitric oxide-induced neuronal injury through the modulation of independent pathways of programmed cell death. J Cereb Blood Flow Metab. 2000;20(9):1380–91. doi: 10.1097/00004647-200009000-00013. [DOI] [PubMed] [Google Scholar]
  • 160.Kelley DE, He J, Menshikova EV, Ritov VB. Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes. Diabetes. 2002;51(10):2944–50. doi: 10.2337/diabetes.51.10.2944. [DOI] [PubMed] [Google Scholar]
  • 161.Choo HJ, Kim JH, Kwon OB, et al. Mitochondria are impaired in the adipocytes of type 2 diabetic mice. Diabetologia. 2006;49(4):784–91. doi: 10.1007/s00125-006-0170-2. [DOI] [PubMed] [Google Scholar]
  • 162.Rex A, Fink H. Pharmacokinetic aspects of reduced nicotinamide adenine dinucleotide (NADH) in rats. Front Biosci. 2008;13:3735–41. doi: 10.2741/2962. [DOI] [PubMed] [Google Scholar]
  • 163.Li F, Chong ZZ, Maiese K. Navigating novel mechanisms of cellular plasticity with the NAD+ precursor and nutrient nicotinamide. Front Biosci. 2004;9:2500–20. doi: 10.2741/1412. [DOI] [PubMed] [Google Scholar]
  • 164.Jackson TM, Rawling JM, Roebuck BD, Kirkland JB. Large supplements of nicotinic acid and nicotinamide increase tissue NAD+ and poly(ADP-ribose) levels but do not affect diethylnitrosamine-induced altered hepatic foci in Fischer-344 rats. J Nutr. 1995;125(6):1455–61. doi: 10.1093/jn/125.6.1455. [DOI] [PubMed] [Google Scholar]
  • 165.Wojcik M, Seidle HF, Bieganowski P, Brenner C. Glutamine-dependent NAD+ synthetase. How a two-domain, three-substrate enzyme avoids waste. J Biol Chem. 2006;281(44):33395–402. doi: 10.1074/jbc.M607111200. [DOI] [PubMed] [Google Scholar]
  • 166.Khan JA, Forouhar F, Tao X, Tong L. Nicotinamide adenine dinucleotide metabolism as an attractive target for drug discovery. Expert opinion on therapeutic targets. 2007;11(5):695–705. doi: 10.1517/14728222.11.5.695. [DOI] [PubMed] [Google Scholar]
  • 167.Khan JA, Xiang S, Tong L. Crystal structure of human nicotinamide riboside kinase. Structure. 2007;15(8):1005–13. doi: 10.1016/j.str.2007.06.017. [DOI] [PubMed] [Google Scholar]
  • 168.Magni G, Amici A, Emanuelli M, et al. Enzymology of NAD+ homeostasis in man. Cell Mol Life Sci. 2004;61(1):19–34. doi: 10.1007/s00018-003-3161-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Lin SJ, Guarente L. Nicotinamide adenine dinucleotide, a metabolic regulator of transcription, longevity and disease. Curr Opin Cell Biol. 2003;15(2):241–6. doi: 10.1016/s0955-0674(03)00006-1. [DOI] [PubMed] [Google Scholar]
  • 170.Hageman GJ, Stierum RH. Niacin, poly(ADP-ribose) polymerase-1 and genomic stability. Mutat Res. 2001;475(1–2):45–56. doi: 10.1016/s0027-5107(01)00078-1. [DOI] [PubMed] [Google Scholar]
  • 171.Idelson M, Alper R, Obolensky A, et al. Directed differentiation of human embryonic stem cells into functional retinal pigment epithelium cells. Cell Stem Cell. 2009;5(4):396–408. doi: 10.1016/j.stem.2009.07.002. [DOI] [PubMed] [Google Scholar]
  • 172.Aoyagi S, Archer TK. Nicotinamide uncouples hormone-dependent chromatin remodeling from transcription complex assembly. Mol Cell Biol. 2008;28(1):30–9. doi: 10.1128/MCB.01158-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Bruno V, Battaglia G, Copani A, et al. Activation of class II or III metabotropic glutamate receptors protects cultured cortical neurons against excitotoxic degeneration. Eur J Neurosci. 1995;7(9):1906–13. doi: 10.1111/j.1460-9568.1995.tb00712.x. [DOI] [PubMed] [Google Scholar]
  • 174.Anderson DW, Bradbury KA, Schneider JS. Neuroprotection in Parkinson models varies with toxin administration protocol. Eur J Neurosci. 2006;24(11):3174–82. doi: 10.1111/j.1460-9568.2006.05192.x. [DOI] [PubMed] [Google Scholar]
  • 175.Chong ZZ, Lin SH, Maiese K. The NAD+ precursor nicotinamide governs neuronal survival during oxidative stress through protein kinase B coupled to FOXO3a and mitochondrial membrane potential. J Cereb Blood Flow Metab. 2004;24(7):728–43. doi: 10.1097/01.WCB.0000122746.72175.0E. [DOI] [PubMed] [Google Scholar]
  • 176.Feng Y, Paul IA, LeBlanc MH. Nicotinamide reduces hypoxic ischemic brain injury in the newborn rat. Brain Res Bull. 2006;69(2):117–22. doi: 10.1016/j.brainresbull.2005.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Lin SH, Chong ZZ, Maiese K. Nicotinamide: a nutritional supplement that provides protection against neuronal and vascular injury. J Med Food. 200;4(1):27–38. doi: 10.1089/10966200152053686. [DOI] [PubMed] [Google Scholar]
  • 178.Slomka M, Zieminska E, Salinska E, Lazarewicz JW. Neuroprotective effects of nicotinamide and 1-methylnicotinamide in acute excitotoxicity in vitro. Folia Neuropathol. 2008;46(1):69–80. [PubMed] [Google Scholar]
  • 179.Slomka M, Zieminska E, Lazarewicz J. Nicotinamide and 1-methylnicotinamide reduce homocysteine neurotoxicity in primary cultures of rat cerebellar granule cells. Acta Neurobiol Exp (Wars) 2008;68(1):1–9. doi: 10.55782/ane-2008-1666. [DOI] [PubMed] [Google Scholar]
  • 180.Ieraci A, Herrera DG. Nicotinamide protects against ethanol-induced apoptotic neurodegeneration in the developing mouse brain. PLoS Med. 2006;3(4):e101. doi: 10.1371/journal.pmed.0030101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Shen CC, Huang HM, Ou HC, et al. Protective effect of nicotinamide on neuronal cells under oxygen and glucose deprivation and hypoxia/reoxygenation. J Biomed Sci. 2004;11(4):472–81. doi: 10.1007/BF02256096. [DOI] [PubMed] [Google Scholar]
  • 182.Sonee M, Martens JR, Evers MR, Mukherjee SK. The effect of tertiary butylhydroperoxide and nicotinamide on human cortical neurons. Neurotoxicology. 2003;24(3):443–8. doi: 10.1016/S0161-813X(03)00019-6. [DOI] [PubMed] [Google Scholar]
  • 183.Kiuchi K, Kondo M, Ueno S, et al. Functional rescue of N-methyl-N-nitrosourea-induced retinopathy by nicotinamide in Sprague-Dawley rats. Curr Eye Res. 2003;26(6):355–62. doi: 10.1076/ceyr.26.5.355.15435. [DOI] [PubMed] [Google Scholar]
  • 184.Kiuchi K, Yoshizawa K, Shikata N, Matsumura M, Tsubura A. Nicotinamide prevents N-methyl-N-nitrosourea-induced photo-receptor cell apoptosis in Sprague-Dawley rats and C57BL mice. Exp Eye Res. 2002;74(3):383–92. doi: 10.1006/exer.2001.1127. [DOI] [PubMed] [Google Scholar]
  • 185.Reber F, Geffarth R, Kasper M, et al. Graded sensitiveness of the various retinal neuron populations on the glyoxal-mediated formation of advanced glycation end products and ways of protection. Graefes Arch Clin Exp Ophthalmol. 2003;241(3):213–25. doi: 10.1007/s00417-002-0528-1. [DOI] [PubMed] [Google Scholar]
  • 186.Hoane MR, Gilbert DR, Holland MA, Pierce JL. Nicotinamide reduces acute cortical neuronal death and edema in the traumatically injured brain. Neurosci Lett. 2006;408(1):35–9. doi: 10.1016/j.neulet.2006.07.011. [DOI] [PubMed] [Google Scholar]
  • 187.Hoane MR, Kaplan SA, Ellis AL. The effects of nicotinamide on apoptosis and blood-brain barrier breakdown following traumatic brain injury. Brain Res. 2006;1125(1):185–93. doi: 10.1016/j.brainres.2006.10.019. [DOI] [PubMed] [Google Scholar]
  • 188.Hoane MR, Pierce JL, Holland MA, Anderson GD. Nicotinamide treatment induces behavioral recovery when administered up to 4 hours following cortical contusion injury in the rat. Neuroscience. 2008;154(3):861–8. doi: 10.1016/j.neuroscience.2008.04.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Hoane MR, Pierce JL, Kaufman NA, Beare JE. Variation in chronic nicotinamide treatment after traumatic brain injury can alter components of functional recovery independent of histological damage. Oxid Med Cell Longev. 2008;1(1):45–52. doi: 10.4161/oxim.1.1.6694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Holland MA, Tan AA, Smith DC, Hoane MR. Nicotinamide treatment provides acute neuroprotection and GFAP regulation following fluid percussion injury. J Neurotrauma. 2008;25(2):140–52. doi: 10.1089/neu.2007.0312. [DOI] [PubMed] [Google Scholar]
  • 191.Wallis RA, Panizzon KL, Girard JM. Traumatic neuroprotection with inhibitors of nitric oxide and ADP- ribosylation. Brain Res. 1996;710(1–2):169–77. doi: 10.1016/0006-8993(95)01278-8. [DOI] [PubMed] [Google Scholar]
  • 192.Wang J, Zhai Q, Chen Y, et al. A local mechanism mediates NAD-dependent protection of axon degeneration. J Cell Biol. 2005;170(3):349–55. doi: 10.1083/jcb.200504028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Yang J, Klaidman L, Chang M, et al. Nicotinamide therapy protects against both necrosis and apoptosis in a stroke model. Pharmacol Biochem Behav. 2002;73(4):901–10. doi: 10.1016/s0091-3057(02)00939-5. [DOI] [PubMed] [Google Scholar]
  • 194.Gupta S, Kaul CL, Sharma SS. Neuroprotective effect of combination of poly (ADP-ribose) polymerase inhibitor and antioxidant in middle cerebral artery occlusion induced focal ischemia in rats. Neurol Res. 2004;26(1):103–7. doi: 10.1179/016164104773026624. [DOI] [PubMed] [Google Scholar]
  • 195.Sakakibara Y, Mitha AP, Ayoub IA, Ogilvy CS, Maynard KI. Delayed treatment with nicotinamide (vitamin B3) reduces the infarct volume following focal cerebral ischemia in spontaneously hypertensive rats, diabetic and non-diabetic Fischer 344 rats. Brain Res. 2002;931(1):68–73. doi: 10.1016/s0006-8993(02)02263-1. [DOI] [PubMed] [Google Scholar]
  • 196.Macleod MR, O’Collins T, Howells DW, Donnan GA. Pooling of animal experimental data reveals influence of study design and publication bias. Stroke. 2004;35(5):1203–8. doi: 10.1161/01.STR.0000125719.25853.20. [DOI] [PubMed] [Google Scholar]
  • 197.Brewer KL, Hardin JS. Neuroprotective effects of nicotinamide after experimental spinal cord injury. Acad Emerg Med. 2004;11(2):125–30. doi: 10.1111/j.1553-2712.2004.tb01421.x. [DOI] [PubMed] [Google Scholar]
  • 198.Isbir CS, Ak K, Kurtkaya O, et al. Ischemic preconditioning and nicotinamide in spinal cord protection in an experimental model of transient aortic occlusion. Eur J Cardiothorac Surg. 2003;23(6):1028–33. doi: 10.1016/s1010-7940(03)00110-6. [DOI] [PubMed] [Google Scholar]
  • 199.Williams A, Ramsden D. Nicotinamide: a double edged sword. Parkinsonism Relat Disord. 2005;11(7):413–20. doi: 10.1016/j.parkreldis.2005.05.011. [DOI] [PubMed] [Google Scholar]
  • 200.Williams AC, Cartwright LS, Ramsden DB. Parkinson’s disease: the first common neurological disease due to auto-intoxication? QJM. 2005;98(3):215–26. doi: 10.1093/qjmed/hci027. [DOI] [PubMed] [Google Scholar]
  • 201.Chong ZZ, Lin SH, Maiese K. Nicotinamide modulates mitochondrial membrane potential and cysteine protease activity during cerebral vascular endothelial cell injury. J Vasc Res. 2002;39(2):131–47. doi: 10.1159/000057762. [DOI] [PubMed] [Google Scholar]
  • 202.Slominska EM, Yuen A, Osman L, et al. Cytoprotective effects of nicotinamide derivatives in endothelial cells. Nucleosides Nucleotides Nucleic Acids. 2008;27(6):863–6. doi: 10.1080/15257770802146528. [DOI] [PubMed] [Google Scholar]
  • 203.Beck H, Plate KH. Angiogenesis after cerebral ischemia. Acta Neuropathol. 2009;117(5):481–96. doi: 10.1007/s00401-009-0483-6. [DOI] [PubMed] [Google Scholar]
  • 204.Giulumian AD, Meszaros LG, Fuchs LC. Endothelin-1-induced contraction of mesenteric small arteries is mediated by ryanodine receptor Ca2+ channels and cyclic ADP-ribose. J Cardiovasc Pharmacol. 2000;36(6):758–63. doi: 10.1097/00005344-200012000-00011. [DOI] [PubMed] [Google Scholar]
  • 205.Pietrzak L, Mogielnicki A, Buczko W. Nicotinamide and its metabolite N-methylnicotinamide increase skin vascular permeability in rats. Clin Exp Dermatol. 2009;34(3):380–4. doi: 10.1111/j.1365-2230.2008.02922.x. [DOI] [PubMed] [Google Scholar]
  • 206.Oumouna-Benachour K, Hans CP, Suzuki Y, et al. Poly(ADP-ribose) polymerase inhibition reduces atherosclerotic plaque size and promotes factors of plaque stability in apolipoprotein E-deficient mice: effects on macrophage recruitment, nuclear factor-kappaB nuclear translocation, and foam cell death. Circulation. 2007;115(18):2442–50. doi: 10.1161/CIRCULATIONAHA.106.668756. [DOI] [PubMed] [Google Scholar]
  • 207.Giammona LM, Fuhrken PG, Papoutsakis ET, Miller WM. Nicotinamide (vitamin B3) increases the polyploidisation and proplatelet formation of cultured primary human megakaryocytes. Br J Haematol. 2006;135(4):554–66. doi: 10.1111/j.1365-2141.2006.06341.x. [DOI] [PubMed] [Google Scholar]
  • 208.Chong ZZ, Lin SH, Li F, Maiese K. The sirtuin inhibitor nicotinamide enhances neuronal cell survival during acute anoxic injury through Akt, Bad, PARP, and mitochondrial associated “anti-apoptotic” pathways. Curr Neurovasc Res. 2005;2(4):271–85. doi: 10.2174/156720205774322584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Maiese K, Lin S, Chong ZZ. Elucidating neuronal and vascular injury through the cytoprotective agent nicotinamide. Curr Med Chem-Immune Endocr Metab Agents. 2001;1(3):257–67. [Google Scholar]
  • 210.Mateuszuk L, Khomich TI, Slominska E, et al. Activation of nicotinamide N-methyltrasferase and increased formation of 1-methylnicotinamide (MNA) in atherosclerosis. Pharmacol Rep. 2009;61(1):76–85. doi: 10.1016/s1734-1140(09)70009-x. [DOI] [PubMed] [Google Scholar]
  • 211.Vincent AM, Maiese K. Nitric oxide induction of neuronal endonuclease activity in programmed cell death. Exp Cell Res. 1999;246(2):290–300. doi: 10.1006/excr.1998.4282. [DOI] [PubMed] [Google Scholar]
  • 212.Vincent AM, TenBroeke M, Maiese K. Metabotropic glutamate receptors prevent programmed cell death through the modulation of neuronal endonuclease activity and intracellular pH. Exp Neurol. 1999;155(1):79–94. doi: 10.1006/exnr.1998.6966. [DOI] [PubMed] [Google Scholar]
  • 213.Vincent AM, TenBroeke M, Maiese K. Neuronal intracellular pH directly mediates nitric oxide-induced programmed cell death. J Neurobiol. 1999;40(2):171–84. doi: 10.1002/(sici)1097-4695(199908)40:2<171::aid-neu4>3.0.co;2-f. [DOI] [PubMed] [Google Scholar]
  • 214.Young GS, Jacobson EL, Kirkland JB. Water maze performance in young male Long-Evans rats is inversely affected by dietary intakes of niacin and may be linked to levels of the NAD+ metabolite cADPR. J Nutr. 2007;137(4):1050–7. doi: 10.1093/jn/137.4.1050. [DOI] [PubMed] [Google Scholar]
  • 215.Reddy S, Bibby NJ, Wu D, et al. A combined casein-free-nicotinamide diet prevents diabetes in the NOD mouse with minimum insulitis. Diabetes Res Clin Pract. 1995;29(2):83–92. doi: 10.1016/0168-8227(95)01109-9. [DOI] [PubMed] [Google Scholar]
  • 216.Hu Y, Wang Y, Wang L, et al. Effects of nicotinamide on prevention and treatment of streptozotocin-induced diabetes mellitus in rats. Chin Med J (Engl) 1996;109(11):819–22. [PubMed] [Google Scholar]
  • 217.Stevens MJ, Li F, Drel VR, et al. Nicotinamide reverses neurological and neurovascular deficits in streptozotocin diabetic rats. J Pharmacol Exp Ther. 2007;320(1):458–64. doi: 10.1124/jpet.106.109702. [DOI] [PubMed] [Google Scholar]
  • 218.Cresto JC, Fabiano de Bruno LE, Cao GF, et al. The association of acetyl-l-carnitine and nicotinamide remits the experimental diabetes in mice by multiple low-dose streptozotocin. Pancreas. 2006;33(4):403–11. doi: 10.1097/01.mpa.0000236740.07854.b1. [DOI] [PubMed] [Google Scholar]
  • 219.Chlopicki S, Swies J, Mogielnicki A, et al. 1-Methylnicotinamide (MNA), a primary metabolite of nicotinamide, exerts anti-thrombotic activity mediated by a cyclooxygenase-2/prostacyclin pathway. Br J Pharmacol. 2007;152(2):230–9. doi: 10.1038/sj.bjp.0707383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Hara N, Yamada K, Shibata T, et al. Elevation of cellular NAD levels by nicotinic acid and involvement of nicotinic acid phosphoribosyltransferase in human cells. J Biol Chem. 2007;282(34):24574–82. doi: 10.1074/jbc.M610357200. [DOI] [PubMed] [Google Scholar]
  • 221.Lee HI, Cho HJ, Han JA, et al. Transient downregulation of protein O-N-acetylglucosaminylation by treatment of high-dose nicotinamide in human cells. Exp Mol Med. 2008;40(2):246–53. doi: 10.3858/emm.2008.40.2.246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Tam D, Tam M, Maynard KI. Nicotinamide modulates energy utilization and improves functional recovery from ischemia in the in vitro rabbit retina. Ann NY Acad Sci. 2005;1053:258–68. doi: 10.1196/annals.1344.023. [DOI] [PubMed] [Google Scholar]
  • 223.Olmos PR, Hodgson MI, Maiz A, et al. Nicotinamide protected first-phase insulin response (FPIR) and prevented clinical disease in first-degree relatives of type-1 diabetics. Diabetes Res Clin Pract. 2006;71(3):320–33. doi: 10.1016/j.diabres.2005.07.009. [DOI] [PubMed] [Google Scholar]
  • 224.Crino A, Schiaffini R, Ciampalini P, et al. A two year observational study of nicotinamide and intensive insulin therapy in patients with recent onset type 1 diabetes mellitus. J Pediatr Endocrinol Metab. 2005;18(8):749–54. doi: 10.1515/jpem.2005.18.8.749. [DOI] [PubMed] [Google Scholar]
  • 225.Eto N, Miyata Y, Ohno H, Yamashita T. Nicotinamide prevents the development of hyperphosphataemia by suppressing intestinal sodium-dependent phosphate transporter in rats with adenine-induced renal failure. Nephrol Dial Transplant. 2005;20(7):1378–84. doi: 10.1093/ndt/gfh781. [DOI] [PubMed] [Google Scholar]
  • 226.Zhou SS, Li D, Sun WP, et al. Nicotinamide overload may play a role in the development of type 2 diabetes. World J Gastroenterol. 2009;15(45):5674–84. doi: 10.3748/wjg.15.5674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Liu HK, Green BD, Flatt PR, McClenaghan NH, McCluskey JT. Effects of long-term exposure to nicotinamide and sodium butyrate on growth, viability, and the function of clonal insulin secreting cells. Endocr Res. 2004;30(1):61–8. doi: 10.1081/erc-120028485. [DOI] [PubMed] [Google Scholar]
  • 228.Reddy S, Salari-Lak N, Sandler S. Long-term effects of nicotinamide-induced inhibition of poly(adenosine diphosphate-ribose) polymerase activity in rat pancreatic islets exposed to interleukin-1 β. Endocrinology. 1995;136(5):1907–12. doi: 10.1210/endo.136.5.7720637. [DOI] [PubMed] [Google Scholar]
  • 229.Gaudineau C, Auclair K. Inhibition of human P450 enzymes by nicotinic acid and nicotinamide. Biochem Biophys Res Commun. 2004;317(3):950–6. doi: 10.1016/j.bbrc.2004.03.137. [DOI] [PubMed] [Google Scholar]
  • 230.Halestrap AP, Woodfield KY, Connern CP. Oxidative stress, thiol reagents, and membrane potential modulate the mitochondrial permeability transition by affecting nucleotide binding to the adenine nucleotide translocase. J Biol Chem. 1997;272(6):3346–54. doi: 10.1074/jbc.272.6.3346. [DOI] [PubMed] [Google Scholar]
  • 231.La Piana G, Marzulli D, Consalvo MI, Lofrumento NE. Cytochrome c-induced cytosolic nicotinamide adenine dinucleotide oxidation, mitochondrial permeability transition, and apoptosis. Arch Biochem Biophys. 2003;410(2):201–11. doi: 10.1016/s0003-9861(02)00687-2. [DOI] [PubMed] [Google Scholar]
  • 232.Maiese K, Li F, Chong ZZ. Erythropoietin in the brain: can the promise to protect be fulfilled? Trends Pharmacol Sci. 2004;25(11):577–83. doi: 10.1016/j.tips.2004.09.006. [DOI] [PubMed] [Google Scholar]
  • 233.Maiese K, Chong Z, Hou J, Shang Y. The “O” class: crafting clinical care with FoxO transcription factors. Adv Exp Med Biol. 2009;665:242–60. doi: 10.1007/978-1-4419-1599-3_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Maiese K, Chong ZZ, Shang YC, Hou J. FoxO proteins: cunning concepts and considerations for the cardiovascular system. Clin Sci (Lond) 2009;116(3):191–203. doi: 10.1042/CS20080113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Maiese K, Hou J, Chong ZZ, Shang YC. A fork in the path: Developing therapeutic inroads with FoxO proteins. Oxid Med Cell Longev. 2009;2(3):119–26. doi: 10.4161/oxim.2.3.8916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Porcu M, Chiarugi A. The emerging therapeutic potential of sirtuin-interacting drugs: from cell death to lifespan extension. Trends Pharmacol Sci. 2005;26(2):94–103. doi: 10.1016/j.tips.2004.12.009. [DOI] [PubMed] [Google Scholar]
  • 237.Saunders LR, Verdin E. Sirtuins: critical regulators at the crossroads between cancer and aging. Oncogene. 2007;26(37):5489–504. doi: 10.1038/sj.onc.1210616. [DOI] [PubMed] [Google Scholar]
  • 238.Maiese K, Chong ZZ, Shang YC. OutFOXOing disease and disability: the therapeutic potential of targeting FoxO proteins. Trends Mol Med. 2008;14(5):219–27. doi: 10.1016/j.molmed.2008.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Taylor DM, Maxwell MM, Luthi-Carter R, Kazantsev AG. Biological and potential therapeutic roles of sirtuin deacetylases. Cell Mol Life Sci. 2008;65(24):4000–18. doi: 10.1007/s00018-008-8357-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Zschoernig B, Mahlknecht U. SIRTUIN 1: regulating the regulator. Biochem Biophys Res Commun. 2008;376(2):251–5. doi: 10.1016/j.bbrc.2008.08.137. [DOI] [PubMed] [Google Scholar]
  • 241.Tang BL, Chua CE. SIRT1 and neuronal diseases. Mol Aspects Med. 2008;29(3):187–200. doi: 10.1016/j.mam.2007.02.001. [DOI] [PubMed] [Google Scholar]
  • 242.Zwaal RF, Schroit AJ. Pathophysiologic implications of membrane phospholipid asymmetry in blood cells. Blood. 1997;89(4):1121–32. [PubMed] [Google Scholar]
  • 243.Canto C, Auwerx J. Caloric restriction, SIRT1 and longevity. Trends Endocrinol Metab. 2009;20(7):325–31. doi: 10.1016/j.tem.2009.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Balan V, Miller GS, Kaplun L, et al. Life span extension and neuronal cell protection by Drosophila nicotinamidase. J Biol Chem. 2008;283(41):27810–9. doi: 10.1074/jbc.M804681200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Bureau G, Longpre F, Martinoli MG. Resveratrol and quercetin, two natural polyphenols, reduce apoptotic neuronal cell death induced by neuroinflammation. J Neurosci Res. 2008;86(2):403–10. doi: 10.1002/jnr.21503. [DOI] [PubMed] [Google Scholar]
  • 246.Chong ZZ, Maiese K. Enhanced tolerance against early and late apoptotic oxidative stress in mammalian neurons through nicotinamidase and sirtuin mediated pathways. Curr Neurovasc Res. 2008;5(3):159–70. doi: 10.2174/156720208785425666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Yoshizaki T, Milne JC, Imamura T, et al. SIRT1 exerts anti-inflammatory effects and improves insulin sensitivity in adipocytes. Mol Cell Biol. 2009;29(5):1363–74. doi: 10.1128/MCB.00705-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Zillikens MC, van Meurs JB, Rivadeneira F, et al. SIRT1 genetic variation is related to BMI and risk of obesity. Diabetes. 2009;58(12):2828–34. doi: 10.2337/db09-0536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Lee HI, Jang SY, Kang HT, Hwang ES. p53-, SIRT1-, and PARP-1-independent downregulation of p21WAF1 expression in nicotinamide-treated cells. Biochem Biophys Res Commun. 2008;368(2):298–304. doi: 10.1016/j.bbrc.2008.01.082. [DOI] [PubMed] [Google Scholar]
  • 250.Jackson MD, Schmidt MT, Oppenheimer NJ, Denu JM. Mechanism of nicotinamide inhibition and transglycosidation by Sir2 histone/protein deacetylases. J Biol Chem. 2003;278(51):50985–98. doi: 10.1074/jbc.M306552200. [DOI] [PubMed] [Google Scholar]
  • 251.Bitterman KJ, Anderson RM, Cohen HY, Latorre-Esteves M, Sinclair DA. Inhibition of silencing and accelerated aging by nicotinamide, a putative negative regulator of yeast sir2 and human SIRT1. J Biol Chem. 2002;277(47):45099–107. doi: 10.1074/jbc.M205670200. [DOI] [PubMed] [Google Scholar]
  • 252.Cai AL, Zipfel GJ, Sheline CT. Zinc neurotoxicity is dependent on intracellular NAD levels and the sirtuin pathway. Eur J Neurosci. 2006;24(8):2169–76. doi: 10.1111/j.1460-9568.2006.05110.x. [DOI] [PubMed] [Google Scholar]
  • 253.Kruszewski M, Szumiel I. Sirtuins (histone deacetylases III) in the cellular response to DNA damage--facts and hypotheses. DNA Repair (Amst) 2005;4(11):1306–13. doi: 10.1016/j.dnarep.2005.06.013. [DOI] [PubMed] [Google Scholar]
  • 254.Maiese K, Chong ZZ, Li F, Shang YC. Erythropoietin: Elucidating new cellular targets that broaden therapeutic strategies. Prog Neurobiol. 2008;85:194–213. doi: 10.1016/j.pneurobio.2008.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Maiese K, Li F, Chong ZZ. New avenues of exploration for erythropoietin. JAMA. 2005;293(1):90–5. doi: 10.1001/jama.293.1.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Girgenti MJ, Hunsberger J, Duman CH, et al. Erythropoietin induction by electroconvulsive seizure, gene regulation, and antidepressant-like behavioral effects. Biol Psychiatry. 2009;66(3):267–74. doi: 10.1016/j.biopsych.2008.12.005. [DOI] [PubMed] [Google Scholar]
  • 257.Ma R, Xiong N, Huang C, et al. Erythropoietin protects PC12 cells from β-amyloid(25–35)-induced apoptosis via PI3K/Akt signaling pathway. Neuropharmacology. 2009;56(6–7):1027–34. doi: 10.1016/j.neuropharm.2009.02.006. [DOI] [PubMed] [Google Scholar]
  • 258.Sun ZK, Yang HQ, Pan J, et al. Protective effects of erythropoietin on tau phosphorylation induced by β-amyloid. J Neurosci Res. 2008;86(13):3018–27. doi: 10.1002/jnr.21745. [DOI] [PubMed] [Google Scholar]
  • 259.McLeod M, Hong M, Mukhida K, et al. Erythropoietin and GDNF enhance ventral mesencephalic fiber outgrowth and capillary proliferation following neural transplantation in a rodent model of Parkinson’s disease. Eur J Neurosci. 2006;24(2):361–70. doi: 10.1111/j.1460-9568.2006.04919.x. [DOI] [PubMed] [Google Scholar]
  • 260.Chong ZZ, Kang JQ, Maiese K. Erythropoietin fosters both intrinsic and extrinsic neuronal protection through modulation of microglia, Akt1, Bad, and caspase-mediated pathways. Br J Pharmacol. 2003;138(6):1107–18. doi: 10.1038/sj.bjp.0705161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Chong ZZ, Maiese K. Erythropoietin involves the phosphatidylino-sitol 3-kinase pathway, 14-3-3 protein and FOXO3a nuclear trafficking to preserve endothelial cell integrity. Br J Pharmacol. 2007;150(7):839–50. doi: 10.1038/sj.bjp.0707161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Cuzzocrea S, Mazzon E, di Paola R, et al. Erythropoietin reduces the degree of arthritis caused by type II collagen in the mouse. Arthritis Rheum. 2005;52(3):940–50. doi: 10.1002/art.20875. [DOI] [PubMed] [Google Scholar]
  • 263.Thorne M, Moore CS, Robertson GS. Lack of TIMP-1 increases severity of experimental autoimmune encephalomyelitis: effects of darbepoetin alfa on TIMP-1 null and wild-type mice. J Neuroimmunol. 2009;211(1–2):92–100. doi: 10.1016/j.jneuroim.2009.04.003. [DOI] [PubMed] [Google Scholar]
  • 264.Sanchez PE, Fares RP, Risso JJ, et al. Optimal neuroprotection by erythropoietin requires elevated expression of its receptor in neurons. Proc Natl Acad Sci USA. 2009;106(24):9848–53. doi: 10.1073/pnas.0901840106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Sanchez PE, Navarro FP, Fares RP, et al. Erythropoietin receptor expression is concordant with erythropoietin but not with common beta chain expression in the rat brain throughout the life span. J Comp Neurol. 2009;514(4):403–14. doi: 10.1002/cne.22020. [DOI] [PubMed] [Google Scholar]
  • 266.Slotkin TA, Seidler FJ, Fumagalli F. Targeting of neurotrophic factors, their receptors, and signaling pathways in the developmental neurotoxicity of organophosphates in vivo and in vitro. Brain Res Bull. 2008;76(4):424–38. doi: 10.1016/j.brainresbull.2008.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.van der Kooij MA, Groenendaal F, Kavelaars A, Heijnen CJ, van Bel F. Neuroprotective properties and mechanisms of erythropoietin in in vitro and in vivo experimental models for hypoxia/ischemia. Brain Res Rev. 2008;59(1):22–33. doi: 10.1016/j.brainresrev.2008.04.007. [DOI] [PubMed] [Google Scholar]
  • 268.Andreotti F, Agati L, Conti E, et al. Update on phase II studies of erythropoietin in acute myocardial infarction. Rationale and design of exogenous erythropoietin in acute myocardial infarction: new outlook and dose association study (EPAMINONDAS) J Thromb Thrombolysis. 2009;28(4):489–95. doi: 10.1007/s11239-009-0363-x. [DOI] [PubMed] [Google Scholar]
  • 269.Brunner S, Winogradow J, Huber BC, et al. Erythropoietin administration after myocardial infarction in mice attenuates ischemic cardiomyopathy associated with enhanced homing of bone marrow-derived progenitor cells via the CXCR-4/SDF-1 axis. FASEB J. 2009;23(2):351–61. doi: 10.1096/fj.08-109462. [DOI] [PubMed] [Google Scholar]
  • 270.Chong ZZ, Kang JQ, Maiese K. Erythropoietin is a novel vascular protectant through activation of Akt1 and mitochondrial modulation of cysteine proteases. Circulation. 2002;106(23):2973–9. doi: 10.1161/01.cir.0000039103.58920.1f. [DOI] [PubMed] [Google Scholar]
  • 271.Incagnoli P, Ramond A, Joyeux-Faure M, et al. Erythropoietin improved initial resuscitation and increased survival after cardiac arrest in rats. Resuscitation. 2009;80(6):696–700. doi: 10.1016/j.resuscitation.2009.03.024. [DOI] [PubMed] [Google Scholar]
  • 272.Li CL, Jiang J, Fan YQ, et al. Knockout of the tumor necrosis factor a receptor 1 gene can up-regulate erythropoietin receptor during myocardial ischemia-reperfusion injury in mice. Chin Med J (Engl) 2009;122(5):566–70. [PubMed] [Google Scholar]
  • 273.Schlecht-Bauer D, Antier D, Machet MC, Hyvelin JM. Short- and long-term cardioprotective effect of darbepoetin-α: role of Bcl-2 family proteins. J Cardiovasc Pharmacol. 2009 Jul 10; doi: 10.1097/FJC.0b013e3181b04d01. [DOI] [PubMed] [Google Scholar]
  • 274.Smith K, Semple D, Bhandari S, Seymour AM. Cellular basis of uraemic cardiomyopathy: a role for erythropoietin? Eur J Heart Fail. 2009;11(8):732–8. doi: 10.1093/eurjhf/hfp093. [DOI] [PubMed] [Google Scholar]
  • 275.Timmer SA, De Boer K, Knaapen P, Gotte MJ, Van Rossum AC. The potential role of erythropoietin in chronic heart failure: from the correction of anemia to improved perfusion and reduced apoptosis? J Card Fail. 2009;15(4):353–61. doi: 10.1016/j.cardfail.2008.10.024. [DOI] [PubMed] [Google Scholar]
  • 276.Uitterdijk A, Groenendijk BC, van Der Giessen WJ. Stem cell therapy for chronic heart failure. Hellenic J Cardiol. 2009;50(2):127–32. [PubMed] [Google Scholar]
  • 277.Matis GK, Birbilis TA. Erythropoietin in spinal cord injury. Eur Spine J. 2009;18(3):314–23. doi: 10.1007/s00586-008-0829-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Yoo JY, Won YJ, Lee JH, et al. Neuroprotective effects of erythropoietin posttreatment against kainate-induced excitotoxicity in mixed spinal cultures. J Neurosci Res. 2009;87(1):150–63. doi: 10.1002/jnr.21832. [DOI] [PubMed] [Google Scholar]
  • 279.Okutan O, Turkoglu OF, Gok HB, Beskonakli E. Neuroprotective effect of erythropoietin after experimental cold injury-induced vasogenic brain edema in rats. Surg Neurol. 2008 Nov;70(5):498–502. doi: 10.1016/j.surneu.2007.07.061. [DOI] [PubMed] [Google Scholar]
  • 280.Karaca M, Odabasoglu F, Kumtepe Y, et al. Protective effects of erythropoietin on ischemia/reperfusion injury of rat ovary. Eur J Obstet Gynecol Reprod Biol. 2009;144(2):157–62. doi: 10.1016/j.ejogrb.2009.03.011. [DOI] [PubMed] [Google Scholar]
  • 281.Harder Y, Amon M, Schramm R, et al. Erythropoietin reduces necrosis in critically ischemic myocutaneous tissue by protecting nutritive perfusion in a dose-dependent manner. Surgery. 2009;145(4):372–83. doi: 10.1016/j.surg.2008.12.001. [DOI] [PubMed] [Google Scholar]
  • 282.Rotter R, Menshykova M, Winkler T, et al. Erythropoietin improves functional and histological recovery of traumatized skeletal muscle tissue. J Orthop Res. 2008 Dec;26(12):1618–26. doi: 10.1002/jor.20692. [DOI] [PubMed] [Google Scholar]
  • 283.Zhu L, Wang HD, Yu XG, et al. Erythropoietin prevents zinc accumulation and neuronal death after traumatic brain injury in rat hippocampus: in vitro and in vivo studies. Brain Res. 2009 Sep 15;1289:96–105. doi: 10.1016/j.brainres.2009.07.015. [DOI] [PubMed] [Google Scholar]
  • 284.Aoshiba K, Onizawa S, Tsuji T, Nagai A. Therapeutic effects of erythropoietin in murine models of endotoxin shock. Crit Care Med. 2009 Mar;37(3):889–988. doi: 10.1097/CCM.0b013e31819b8371. [DOI] [PubMed] [Google Scholar]
  • 285.Contaldo C, Elsherbiny A, Lindenblatt N, et al. Erythropoietin enhances oxygenation in critically perfused tissue through modulation of nitric oxide synthase. Shock. 2009;31(6):599–606. doi: 10.1097/SHK.0b013e31818b9cc4. [DOI] [PubMed] [Google Scholar]
  • 286.Simon F, Calzia E, Radermacher P, Schelzig H. Beneficial effects of erythropoietin in models of shock and organ failure-nothing is simple and easy. Shock. 2009;31(2):220–1. doi: 10.1097/SHK.0b013e3181890733. [DOI] [PubMed] [Google Scholar]
  • 287.Casals-Pascual C, Idro R, Picot S, Roberts DJ, Newton CR. Can erythropoietin be used to prevent brain damage in cerebral malaria? Trends Parasitol. 2009;25(1):30–6. doi: 10.1016/j.pt.2008.10.002. [DOI] [PubMed] [Google Scholar]
  • 288.Mihaila RG, Rezi EC, Boitan M, et al. Erythropoietin and the pro-inflammatory cytokines in chronic C hepatitis. Hepatogastroenterology. 2009;56(91–92):751–5. [PubMed] [Google Scholar]
  • 289.Picot S, Bienvenu AL, Konate S, et al. Safety of epoietin beta-quinine drug combination in children with cerebral malaria in Mali. Malar J. 2009;8:169. doi: 10.1186/1475-2875-8-169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.MacRedmond R, Singhera GK, Dorscheid DR. Erythropoietin inhibits respiratory epithelial cell apoptosis in a model of acute lung injury. Eur Respir J. 2009;33(6):1403–14. doi: 10.1183/09031936.00084608. [DOI] [PubMed] [Google Scholar]
  • 291.Tascilar O, Cakmak GK, Tekin IO, et al. Protective effects of erythropoietin against acute lung injury in a rat model of acute necrotizing pancreatitis. World J Gastroenterol. 2007;13(46):6172–82. doi: 10.3748/wjg.v13.i46.6172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Wu H, Dong G, Liu H, et al. Erythropoietin attenuates ischemia-reperfusion induced lung injury by inhibiting tumor necrosis factor-alpha and matrix metalloproteinase-9 expression. Eur J Pharmacol. 2009;602(2–3):406–12. doi: 10.1016/j.ejphar.2008.11.037. [DOI] [PubMed] [Google Scholar]
  • 293.Chang YK, Choi DE, Na KR, et al. Erythropoietin attenuates renal injury in an experimental model of rat unilateral ureteral obstruction via anti-inflammatory and anti-apoptotic effects. J Urol. 2009;181(3):1434–43. doi: 10.1016/j.juro.2008.10.105. [DOI] [PubMed] [Google Scholar]
  • 294.Chen HH, Tarng DC, Lee KF, Wu CY, Chen YC. Epoetin alfa and darbepoetin alfa: effects on ventricular hypertrophy in patients with chronic kidney disease. J Nephrol. 2008;21(4):543–9. [PubMed] [Google Scholar]
  • 295.Song YR, Lee T, You SJ, et al. Prevention of acute kidney injury by erythropoietin in patients undergoing coronary artery bypass grafting: a pilot study. Am J Nephrol. 2009;30(3):253–60. doi: 10.1159/000223229. [DOI] [PubMed] [Google Scholar]
  • 296.Luo YH, Li ZD, Liu LX, Dong GH. Pretreatment with erythropoietin reduces hepatic ischemia-reperfusion injury. Hepatobiliary Pancreat Dis Int. 2009;8(3):294–9. [PubMed] [Google Scholar]
  • 297.Schmeding M, Hunold G, Ariyakhagorn V, et al. Erythropoietin reduces ischemia-reperfusion injury after liver transplantation in rats. Transpl Int. 2009;22(7):738–46. doi: 10.1111/j.1432-2277.2009.00861.x. [DOI] [PubMed] [Google Scholar]
  • 298.Ucan BH, Irkorucu O, Cakmak GK, et al. Erythropoietin: a possible cytoprotective cytokine in acute necrotizing pancreatitis. J Hepatobiliary Pancreat Surg. 2009;16(4):530–7. doi: 10.1007/s00534-009-0082-x. [DOI] [PubMed] [Google Scholar]
  • 299.Lagreze WA, Feltgen N, Bach M, Jehle T. Feasibility of intravitreal erythropoietin injections in humans. Br J Ophthalmol. 2009;93(12):1667–71. doi: 10.1136/bjo.2008.156794. [DOI] [PubMed] [Google Scholar]
  • 300.Tsai JC, Song BJ, Wu L, Forbes M. Erythropoietin: a candidate neuroprotective agent in the treatment of glaucoma. J Glaucoma. 2007;16(6):567–71. doi: 10.1097/IJG.0b013e318156a556. [DOI] [PubMed] [Google Scholar]
  • 301.Wang ZY, Shen LJ, Tu L, et al. Erythropoietin protects retinal pigment epithelial cells from oxidative damage. Free Radic Biol Med. 2009;46(8):1032–41. doi: 10.1016/j.freeradbiomed.2008.11.027. [DOI] [PubMed] [Google Scholar]
  • 302.Zhong YS, Liu XH, Cheng Y, Min YJ. Erythropoietin with retrobulbar administration protects retinal ganglion cells from acute elevated intraocular pressure in rats. J Ocul Pharmacol Ther. 2008;24(5):453–9. doi: 10.1089/jop.2008.0021. [DOI] [PubMed] [Google Scholar]
  • 303.Toba H, Sawai N, Morishita M, et al. Chronic treatment with recombinant human erythropoietin exerts renoprotective effects beyond hematopoiesis in streptozotocin-induced diabetic rat. Eur J Pharmacol. 2009;612(1–3):106–14. doi: 10.1016/j.ejphar.2009.03.065. [DOI] [PubMed] [Google Scholar]
  • 304.Mikati MA, Hokayem JA, Sabban ME. Effects of a single dose of erythropoietin on subsequent seizure susceptibility in rats exposed to acute hypoxia at p10. Epilepsia. 2007;48(1):175–81. doi: 10.1111/j.1528-1167.2006.00900.x. [DOI] [PubMed] [Google Scholar]
  • 305.Moon C, Krawczyk M, Paik D, et al. Erythropoietin, modified to not stimulate red blood cell production, retains its cardioprotective properties. J Pharmacol Exp Ther. 2006;316(3):999–1005. doi: 10.1124/jpet.105.094854. [DOI] [PubMed] [Google Scholar]
  • 306.Um M, Gross AW, Lodish HF. A “classical” homodimeric erythropoietin receptor is essential for the antiapoptotic effects of erythropoietin on differentiated neuroblastoma SH-SY5Y and pheochromocytoma PC-12 cells. Cell Signal. 2007;19(3):634–45. doi: 10.1016/j.cellsig.2006.08.014. [DOI] [PubMed] [Google Scholar]
  • 307.Di Giacomo V, Sancilio S, Caravatta L, et al. Regulation of CREB activation by P38 mitogen activated protein kinase during human primary erythroblast differentiation. Int J Immunopathol Pharmacol. 2009;22(3):679–88. doi: 10.1177/039463200902200313. [DOI] [PubMed] [Google Scholar]
  • 308.Koh SH, Noh MY, Cho GW, Kim KS, Kim SH. Erythropoietin increases the motility of human bone marrow-multipotent stromal cells (hBM-MSCs) and enhances the production of neurotrophic factors from hBM-MSCs. Stem Cells Dev. 2009;18(3):411–21. doi: 10.1089/scd.2008.0040. [DOI] [PubMed] [Google Scholar]
  • 309.Tilling L, Chowienczyk P, Clapp B. Progenitors in motion: mechanisms of mobilization of endothelial progenitor cells. Br J Clin Pharmacol. 2009;68(4):484–92. doi: 10.1111/j.1365-2125.2009.03486.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Arcasoy MO. The non-haematopoietic biological effects of erythropoietin. Br J Haematol. 2008;141(1):14–31. doi: 10.1111/j.1365-2141.2008.07014.x. [DOI] [PubMed] [Google Scholar]
  • 311.Schumann C, Triantafilou K, Krueger S, et al. Detection of erythropoietin in exhaled breath condensate of nonhypoxic subjects using a multiplex bead array. Mediators Inflamm. 2006;2006(5):18061. doi: 10.1155/MI/2006/18061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312.Li F, Chong ZZ, Maiese K. Erythropoietin on a tightrope: balancing neuronal and vascular protection between intrinsic and extrinsic pathways. Neurosignals. 2004;13(6):265–89. doi: 10.1159/000081963. [DOI] [PubMed] [Google Scholar]
  • 313.Maiese K, Chong ZZ, Shang YC. Raves and risks for erythropoietin. Cytokine Growth Factor Rev. 2008;19(2):145–55. doi: 10.1016/j.cytogfr.2008.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314.Mojiminiyi OA, Abdella NA, Zaki MY, et al. Prevalence and associations of low plasma erythropoietin in patients with Type 2 diabetes mellitus. Diabet Med. 2006;23(8):839–44. doi: 10.1111/j.1464-5491.2006.01893.x. [DOI] [PubMed] [Google Scholar]
  • 315.Symeonidis A, Kouraklis-Symeonidis A, Psiroyiannis A, et al. Inappropriately low erythropoietin response for the degree of anemia in patients with noninsulin-dependent diabetes mellitus. Ann Hematol. 2006;85(2):79–85. doi: 10.1007/s00277-005-1102-9. [DOI] [PubMed] [Google Scholar]
  • 316.Thomas MC, Cooper ME, Tsalamandris C, MacIsaac R, Jerums G. Anemia with impaired erythropoietin response in diabetic patients. Arch Intern Med. 2005;28; 165(4):466–9. doi: 10.1001/archinte.165.4.466. [DOI] [PubMed] [Google Scholar]
  • 317.Teramo K, Kari MA, Eronen M, Markkanen H, Hiilesmaa V. High amniotic fluid erythropoietin levels are associated with an increased frequency of fetal and neonatal morbidity in type 1 diabetic pregnancies. Diabetologia. 2004;47(10):1695–703. doi: 10.1007/s00125-004-1515-3. [DOI] [PubMed] [Google Scholar]
  • 318.Teramo KA, Widness JA. Increased fetal plasma and amniotic fluid erythropoietin concentrations: markers of intrauterine hypoxia. Neonatology. 2009;95(2):105–16. doi: 10.1159/000153094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 319.Silverberg DS, Wexler D, Iaina A, Schwartz D. The interaction between heart failure and other heart diseases, renal failure, and anemia. Semin Nephrol. 2006;26(4):296–306. doi: 10.1016/j.semnephrol.2006.05.006. [DOI] [PubMed] [Google Scholar]
  • 320.Bierer R, Peceny MC, Hartenberger CH, Ohls RK. Erythropoietin concentrations and neurodevelopmental outcome in preterm infants. Pediatrics. 2006;118(3):e635–40. doi: 10.1542/peds.2005-3186. [DOI] [PubMed] [Google Scholar]
  • 321.Pillai A, Dhandapani KM, Pillai BA, Terry AV, Jr, Mahadik SP. Erythropoietin prevents haloperidol treatment-induced neuronal apoptosis through regulation of BDNF. Neuropsychopharmacology. 2008;33(8):1942–51. doi: 10.1038/sj.npp.1301566. [DOI] [PubMed] [Google Scholar]
  • 322.Kaindl AM, Sifringer M, Koppelstaetter A, et al. Erythropoietin protects the developing brain from hyperoxia-induced cell death and proteome changes. Ann Neurol. 2008;64(5):523–34. doi: 10.1002/ana.21471. [DOI] [PubMed] [Google Scholar]
  • 323.Yis U, Kurul SH, Kumral A, et al. Effect of erythropoietin on oxygen-induced brain injury in the newborn rat. Neurosci Lett. 2008;448(3):245–9. doi: 10.1016/j.neulet.2008.10.060. [DOI] [PubMed] [Google Scholar]
  • 324.He Z, Huang L, Wu Y, et al. DDPH: improving cognitive deficits beyond its α 1-adrenoceptor antagonism in chronic cerebral hypoperfused rats. Eur J Pharmacol. 2008;588(2–3):178–88. doi: 10.1016/j.ejphar.2008.03.060. [DOI] [PubMed] [Google Scholar]
  • 325.Berkingali N, Warnecke A, Gomes P, et al. Neurite outgrowth on cultured spiral ganglion neurons induced by erythropoietin. Hear Res. 2008;243(1–2):121–6. doi: 10.1016/j.heares.2008.07.003. [DOI] [PubMed] [Google Scholar]
  • 326.Shah RC, Wilson RS, Tang Y, et al. Relation of hemoglobin to level of cognitive function in older persons. Neuroepidemiology. 2009;32(1):40–6. doi: 10.1159/000170905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 327.Mason-Garcia M, Beckman BS, Brookins JW, et al. Development of a new radioimmunoassay for erythropoietin using recombinant erythropoietin. Kidney Int. 1990;38(5):969–75. doi: 10.1038/ki.1990.299. [DOI] [PubMed] [Google Scholar]
  • 328.Namiuchi S, Kagaya Y, Ohta J, et al. High serum erythropoietin level is associated with smaller infarct size in patients with acute myocardial infarction who undergo successful primary percutaneous coronary intervention. J Am Coll Cardiol. 2005;45(9):1406–12. doi: 10.1016/j.jacc.2005.01.043. [DOI] [PubMed] [Google Scholar]
  • 329.Gayer CP, Chaturvedi LS, Wang S, et al. Strain-induced proliferation requires the phosphatidylinositol 3-kinase/AKT/glycogen synthase kinase pathway. J Biol Chem. 2009;284(4):2001–11. doi: 10.1074/jbc.M804576200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.An J, Zhang C, Polavarapu R, Zhang X, Yepes M. Tissue-type plasminogen activator and the low-density lipoprotein receptor-related protein induce Akt phosphorylation in the ischemic brain. Blood. 2008;112(7):2787–94. doi: 10.1182/blood-2008-02-141630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 331.Slaets H, Dumont D, Vanderlocht J, et al. Leukemia inhibitory factor induces an antiapoptotic response in oligodendrocytes through Akt-phosphorylation and up-regulation of 14–3-3. Proteomics. 2008;8(6):1237–47. doi: 10.1002/pmic.200700641. [DOI] [PubMed] [Google Scholar]
  • 332.Dreixler JC, Hemmert JW, Shenoy SK, et al. The role of Akt/protein kinase B subtypes in retinal ischemic preconditioning. Exp Eye Res. 2009;88(3):512–21. doi: 10.1016/j.exer.2008.11.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 333.Anitha M, Gondha C, Sutliff R, et al. GDNF rescues hyperglycemia-induced diabetic enteric neuropathy through activation of the PI3K/Akt pathway. J Clin Invest. 2006;116(2):344–56. doi: 10.1172/JCI26295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 334.Burgos-Ramos E, Martos-Moreno GA, Lopez MG, et al. The N-terminal tripeptide of insulin-like growth factor-I protects against beta-amyloid-induced somatostatin depletion by calcium and glycogen synthase kinase 3 β modulation. J Neurochem. 2009;109(2):360–70. doi: 10.1111/j.1471-4159.2009.05980.x. [DOI] [PubMed] [Google Scholar]
  • 335.Tamagno E, Guglielmotto M, Giliberto L, et al. JNK and ERK1/2 pathways have a dual opposite effect on the expression of BACE1. Neurobiol Aging. 2009;30(10):1563–73. doi: 10.1016/j.neurobiolaging.2007.12.015. [DOI] [PubMed] [Google Scholar]
  • 336.Kim KH, Oudit GY, Backx PH. Erythropoietin protects against doxorubicin-induced cardiomyopathy via a phosphatidylinositol 3-kinase-dependent pathway. J Pharmacol Exp Ther. 2008;324(1):160–9. doi: 10.1124/jpet.107.125773. [DOI] [PubMed] [Google Scholar]
  • 337.Tajes M, Yeste-Velasco M, Zhu X, et al. Activation of Akt by lithium: pro-survival pathways in aging. Mech Ageing Dev. 2009;130(4):253–61. doi: 10.1016/j.mad.2008.12.006. [DOI] [PubMed] [Google Scholar]
  • 338.Chong ZZ, Kang JQ, Maiese K. Akt1 drives endothelial cell membrane asymmetry and microglial activation through Bcl-x(L) and caspase 1, 3, and 9. Exp Cell Res. 2004;296(2):196–207. doi: 10.1016/j.yexcr.2004.01.021. [DOI] [PubMed] [Google Scholar]
  • 339.Fujiwara M, Izuishi K, Sano T, et al. Modulating effect of the PI3-kinase inhibitor LY294002 on cisplatin in human pancreatic cancer cells. J Exp Clin Cancer Res. 2008;27:76. doi: 10.1186/1756-9966-27-76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 340.Bahlmann FH, Song R, Boehm SM, et al. Low-dose therapy with the long-acting erythropoietin analogue darbepoetin α persistently activates endothelial Akt and attenuates progressive organ failure. Circulation. 2004;110(8):1006–12. doi: 10.1161/01.CIR.0000139335.04152.F3. [DOI] [PubMed] [Google Scholar]
  • 341.Miki T, Miura T, Yano T, et al. Alteration in erythropoietin-induced cardioprotective signaling by postinfarct ventricular remodeling. J Pharmacol Exp Ther. 2006;317(1):68–75. doi: 10.1124/jpet.105.095745. [DOI] [PubMed] [Google Scholar]
  • 342.Parsa CJ, Matsumoto A, Kim J, et al. A novel protective effect of erythropoietin in the infarcted heart. J Clin Invest. 2003;112(7):999–1007. doi: 10.1172/JCI18200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 343.Sharples EJ, Patel N, Brown P, et al. Erythropoietin protects the kidney against the injury and dysfunction caused by ischemia-reperfusion. J Am Soc Nephrol. 2004;15(8):2115–24. doi: 10.1097/01.ASN.0000135059.67385.5D. [DOI] [PubMed] [Google Scholar]
  • 344.Um M, Lodish HF. Antiapoptotic effects of erythropoietin in differentiated neuroblastoma SH-SY5Y cells require activation of both the STAT5 and AKT signaling pathways. J Biol Chem. 2006;281(9):5648–56. doi: 10.1074/jbc.M510943200. [DOI] [PubMed] [Google Scholar]
  • 345.Wu Y, Shang Y, Sun S, Liu R. Antioxidant effect of erythropoietin on 1-methyl-4-phenylpyridinium-induced neurotoxicity in PC12 cells. Eur J Pharmacol. 2007;564(1–3):47–56. doi: 10.1016/j.ejphar.2007.02.020. [DOI] [PubMed] [Google Scholar]
  • 346.Matsuzaki H, Tamatani M, Mitsuda N, et al. Activation of Akt kinase inhibits apoptosis and changes in Bcl-2 and Bax expression induced by nitric oxide in primary hippocampal neurons. J Neurochem. 1999;73(5):2037–46. [PubMed] [Google Scholar]
  • 347.Rytomaa M, Lehmann K, Downward J. Matrix detachment induces caspase-dependent cytochrome c release from mitochondria: inhibition by PKB/Akt but not Raf signalling. Oncogene. 2000;19(39):4461–8. doi: 10.1038/sj.onc.1203805. [DOI] [PubMed] [Google Scholar]
  • 348.Namikawa K, Honma M, Abe K, et al. Akt/protein kinase B prevents injury-induced motoneuron death and accelerates axonal regeneration. J Neurosci. 2000;20(8):2875–86. doi: 10.1523/JNEUROSCI.20-08-02875.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 349.Dzietko M, Felderhoff-Mueser U, Sifringer M, et al. Erythropoietin protects the developing brain against N-methyl-D-aspartate receptor antagonist neurotoxicity. Neurobiol Dis. 2004;15(2):177–87. doi: 10.1016/j.nbd.2003.10.006. [DOI] [PubMed] [Google Scholar]
  • 350.Zhang Y, Park TS, Gidday JM. Hypoxic preconditioning protects human brain endothelium from ischemic apoptosis by Akt-dependent survivin activation. Am J Physiol Heart Circ Physiol. 2007;292(6):H2573–81. doi: 10.1152/ajpheart.01098.2006. [DOI] [PubMed] [Google Scholar]
  • 351.Martin D, Salinas M, Lopez-Valdaliso R, et al. Effect of the Alzheimer amyloid fragment Aβ(25–35) on Akt/PKB kinase and survival of PC12 cells. J Neurochem. 2001;78(5):1000–8. doi: 10.1046/j.1471-4159.2001.00472.x. [DOI] [PubMed] [Google Scholar]
  • 352.Henry MK, Lynch JT, Eapen AK, Quelle FW. DNA damage-induced cell-cycle arrest of hematopoietic cells is overridden by activation of the PI-3 kinase/Akt signaling pathway. Blood. 2001;98(3):834–41. doi: 10.1182/blood.v98.3.834. [DOI] [PubMed] [Google Scholar]
  • 353.Maiese K, Chong ZZ, Li F. Driving cellular plasticity and survival through the signal transduction pathways of metabotropic glutamate receptors. Curr Neurovasc Res. 2005;2(5):425–46. doi: 10.2174/156720205774962692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 354.Maiese K, Chong ZZ, Shang YC. “Sly as a FOXO”: New paths with Forkhead signaling in the brain. Curr Neurovasc Res. 2007;4(4):295–302. doi: 10.2174/156720207782446306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 355.Coffer PJ. When less is more: the PI3K pathway as a determinant of tumor response to dietary restriction. Cell Res. 2009;19(7):797–9. doi: 10.1038/cr.2009.81. [DOI] [PubMed] [Google Scholar]
  • 356.Jacobsen EA, Ananieva O, Brown ML, Chang Y. Growth, differentiation, and malignant transformation of pre-B cells mediated by inducible activation of v-Abl oncogene. J Immunol. 2006;176(11):6831–8. doi: 10.4049/jimmunol.176.11.6831. [DOI] [PubMed] [Google Scholar]
  • 357.Maiese K, Chong ZZ, Shang YC, Hou J. Clever cancer strategies with FoxO transcription factors. Cell Cycle. 2008;7(24):3829–39. doi: 10.4161/cc.7.24.7231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 358.Kaestner KH, Knochel W, Martinez DE. Unified nomenclature for the winged helix/forkhead transcription factors. Genes Dev. 2000;14(2):142–6. [PubMed] [Google Scholar]
  • 359.Lappas M, Lim R, Riley C, Rice GE, Permezel M. Localisation and expression of FoxO1 proteins in human gestational tissues. Placenta. 2009;30(3):256–62. doi: 10.1016/j.placenta.2008.12.008. [DOI] [PubMed] [Google Scholar]
  • 360.Zheng WH, Kar S, Quirion R. FKHRL1 and its homologs are new targets of nerve growth factor Trk receptor signaling. J Neurochem. 2002;80(6):1049–61. doi: 10.1046/j.0022-3042.2002.00783.x. [DOI] [PubMed] [Google Scholar]
  • 361.Fei M, Lu M, Wang Y, et al. Arsenic trioxide-induced growth arrest of human hepatocellular carcinoma cells involving FOXO3a expression and localization. Med Oncol. 2009;26(2):178–85. doi: 10.1007/s12032-008-9105-8. [DOI] [PubMed] [Google Scholar]
  • 362.Cariou A, Claessens YE, Pene F, et al. Early high-dose erythropoietin therapy and hypothermia after out-of-hospital cardiac arrest: a matched control study. Resuscitation. 2008;76(3):397–404. doi: 10.1016/j.resuscitation.2007.10.003. [DOI] [PubMed] [Google Scholar]
  • 363.Bakker WJ, van Dijk TB, Parren-van Amelsvoort M, et al. Differential regulation of Foxo3a target genes in erythropoiesis. Mol Cell Biol. 2007;27(10):3839–54. doi: 10.1128/MCB.01662-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 364.Lei H, Quelle FW. FOXO transcription factors enforce cell cycle checkpoints and promote survival of hematopoietic cells after DNA damage. Mol Cancer Res. 2009;7(8):1294–303. doi: 10.1158/1541-7786.MCR-08-0531. [DOI] [PubMed] [Google Scholar]
  • 365.Bethea CL, Reddy AP, Tokuyama Y, Henderson JA, Lima FB. Protective actions of ovarian hormones in the serotonin system of macaques. Front Neuroendocrinol. 2009;30(2):212–38. doi: 10.1016/j.yfrne.2009.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366.Chong ZZ, Kang JQ, Maiese K. Essential cellular regulatory elements of oxidative stress in early and late phases of apoptosis in the central nervous system. Antioxid Redox Signal. 2004;6(2):277–87. doi: 10.1089/152308604322899341. [DOI] [PubMed] [Google Scholar]
  • 367.Fallarino F, Bianchi R, Orabona C, et al. CTLA-4-Ig activates forkhead transcription factors and protects dendritic cells from oxidative stress in nonobese diabetic mice. J Exp Med. 2004;200(8):1051–62. doi: 10.1084/jem.20040942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 368.Nakae J, Cao Y, Oki M, et al. Forkhead transcription factor FoxO1 in adipose tissue regulates energy storage and expenditure. Diabetes. 2008;57(3):563–76. doi: 10.2337/db07-0698. [DOI] [PubMed] [Google Scholar]
  • 369.Furuyama T, Yamashita H, Kitayama K, et al. Effects of aging and caloric restriction on the gene expression of Foxo1, 3, and 4 (FKHR, FKHRL1, and AFX) in the rat skeletal muscles. Microsc Res Tech. 2002;59(4):331–4. doi: 10.1002/jemt.10213. [DOI] [PubMed] [Google Scholar]
  • 370.Puig O, Tjian R. Transcriptional feedback control of insulin receptor by dFOXO/FOXO1. Genes Dev. 2005;19(20):2435–46. doi: 10.1101/gad.1340505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 371.Kim JR, Jung HS, Bae SW, et al. Polymorphisms in FOXO gene family and association analysis with BMI. Obesity (Silver Spring, MD) 2006;14(2):188–93. doi: 10.1038/oby.2006.24. [DOI] [PubMed] [Google Scholar]
  • 372.Marchetti V, Menghini R, Rizza S, et al. Benfotiamine counteracts glucose toxicity effects on endothelial progenitor cell differentiation via Akt/FoxO signaling. Diabetes. 2006;55(8):2231–7. doi: 10.2337/db06-0369. [DOI] [PubMed] [Google Scholar]
  • 373.Kamagate A, Dong HH. Foxo1 integrates insulin signaling to VLDL production. Cell Cycle. 2008;7(20):3162–70. doi: 10.4161/cc.7.20.6882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 374.Ni YG, Wang N, Cao DJ, et al. FoxO transcription factors activate Akt and attenuate insulin signaling in heart by inhibiting protein phosphatases. Proc Natl Acad Sci USA. 2007;104(51):20517–22. doi: 10.1073/pnas.0610290104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 375.Kamei Y, Miura S, Suzuki M, et al. Skeletal muscle FOXO1 (FKHR) transgenic mice have less skeletal muscle mass, down-regulated Type I (slow twitch/red muscle) fiber genes, and impaired glycemic control. J Biol Chem. 2004;279(39):41114–23. doi: 10.1074/jbc.M400674200. [DOI] [PubMed] [Google Scholar]
  • 376.Liu CM, Yang Z, Liu CW, et al. Effect of RNA oligonucleotide targeting Foxo-1 on muscle growth in normal and cancer cachexia mice. Cancer Gene Ther. 2007;14(12):945–52. doi: 10.1038/sj.cgt.7701091. [DOI] [PubMed] [Google Scholar]
  • 377.Sandri M, Lin J, Handschin C, et al. PGC-1α protects skeletal muscle from atrophy by suppressing FoxO3 action and atrophy-specific gene transcription. Proc Natl Acad Sci USA. 2006;103(44):16260–5. doi: 10.1073/pnas.0607795103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 378.Maiese K, Li F, Chong ZZ, Shang YC. The Wnt signaling pathway: Aging gracefully as a protectionist? Pharmacol Ther. 2008;118(1):58–81. doi: 10.1016/j.pharmthera.2008.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 379.Speese SD, Budnik V. Wnts: up-and-coming at the synapse. Trends Neurosci. 2007;30(6):268–75. doi: 10.1016/j.tins.2007.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 380.Espada J, Calvo MB, Diaz-Prado S, Medina V. Wnt signalling and cancer stem cells. Clin Transl Oncol. 2009 Jul;11(7):411–27. doi: 10.1007/s12094-009-0380-4. [DOI] [PubMed] [Google Scholar]
  • 381.Jozwiak J, Kotulska K, Grajkowska W, et al. Upregulation of the WNT pathway in tuberous sclerosis-associated subependymal giant cell astrocytomas. Brain Dev. 2007;29(5):273–80. doi: 10.1016/j.braindev.2006.09.009. [DOI] [PubMed] [Google Scholar]
  • 382.Jozwiak J, Wlodarski P. Hamartin and tuberin modulate gene transcription via β-catenin. J Neurooncol. 2006;79(3):229–34. doi: 10.1007/s11060-006-9134-0. [DOI] [PubMed] [Google Scholar]
  • 383.Kikuchi A, Yamamoto H, Sato A. Selective activation mechanisms of Wnt signaling pathways. Trends Cell Biol. 2009;19(3):119–29. doi: 10.1016/j.tcb.2009.01.003. [DOI] [PubMed] [Google Scholar]
  • 384.Lee JH, Lee EO, Kang JL, Chong YH. Concomitant degradation of beta-catenin and GSK-3 β potently contributes to glutamate-induced neurotoxicity in rat hippocampal slice cultures. J Neurochem. 2008;106(3):1066–77. doi: 10.1111/j.1471-4159.2008.05444.x. [DOI] [PubMed] [Google Scholar]
  • 385.Luo JM, Dai CF, Lin SY, Huang PQ. Asymmetric syntheses and Wnt signal inhibitory activity of melleumin A and four analogues of melleumins A and B. Chem Asian J. 2009;4(2):328–35. doi: 10.1002/asia.200800355. [DOI] [PubMed] [Google Scholar]
  • 386.Mercado-Gomez O, Hernandez-Fonseca K, Villavicencio-Queijeiro A, et al. Inhibition of Wnt and PI3K signaling modulates GSK-3beta activity and induces morphological changes in cortical neurons: role of tau phosphorylation. Neurochem Res. 2008;33(8):1599–609. doi: 10.1007/s11064-008-9714-9. [DOI] [PubMed] [Google Scholar]
  • 387.Muruganandan S, Roman AA, Sinal CJ. Adipocyte differentiation of bone marrow-derived mesenchymal stem cells: cross talk with the osteoblastogenic program. Cell Mol Life Sci. 2009;66(2):236–53. doi: 10.1007/s00018-008-8429-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 388.Sutton LP, Honardoust D, Mouyal J, Rajakumar N, Rushlow WJ. Activation of the canonical Wnt pathway by the antipsychotics haloperidol and clozapine involves dishevelled-3. J Neurochem. 2007;102(1):153–69. doi: 10.1111/j.1471-4159.2007.04527.x. [DOI] [PubMed] [Google Scholar]
  • 389.Vettor R, Milan G, Franzin C, et al. The origin of intermuscular adipose tissue and its pathophysiological implications. Am J Physiol Endocrinol Metab. 2009 Sep 8; doi: 10.1152/ajpendo.00229.2009. [DOI] [PubMed] [Google Scholar]
  • 390.Wang Z, Havasi A, Gall JM, et al. β-catenin promotes survival of renal epithelial cells by inhibiting Bax. J Am Soc Nephrol. 2009;20(9):1919–28. doi: 10.1681/ASN.2009030253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 391.Wilusz M, Majka M. Role of the Wnt/β-catenin network in regulating hematopoiesis. Arch Immunol Ther Exp (Warsz) 2008;56(4):257–66. doi: 10.1007/s00005-008-0029-y. [DOI] [PubMed] [Google Scholar]
  • 392.Grant SF, Thorleifsson G, Reynisdottir I, et al. Variant of transcription factor 7-like 2 (TCF7L2) gene confers risk of type 2 diabetes. Nat Genet. 2006;38(3):320–3. doi: 10.1038/ng1732. [DOI] [PubMed] [Google Scholar]
  • 393.Lehman DM, Hunt KJ, Leach RJ, et al. Haplotypes of transcription factor 7-like 2 (TCF7L2) gene and its upstream region are associated with type 2 diabetes and age of onset in mexican americans. Diabetes. 2007;56(2):389–93. doi: 10.2337/db06-0860. [DOI] [PubMed] [Google Scholar]
  • 394.Scott LJ, Bonnycastle LL, Willer CJ, et al. Association of transcription factor 7-like 2 (TCF7L2) variants with type 2 diabetes in a Finnish sample. Diabetes. 2006;55(9):2649–53. doi: 10.2337/db06-0341. [DOI] [PubMed] [Google Scholar]
  • 395.Guo YF, Xiong DH, Shen H, et al. Polymorphisms of the low-density lipoprotein receptor-related protein 5 (LRP5) gene are associated with obesity phenotypes in a large family-based association study. J Med Genet. 2006;43(10):798–803. doi: 10.1136/jmg.2006.041715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 396.Kanazawa A, Tsukada S, Sekine A, et al. Association of the gene encoding wingless-type mammary tumor virus integration-site family member 5B (WNT5B) with type 2 diabetes. Am J Hum Genet. 2004;75(5):832–43. doi: 10.1086/425340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 397.Al-Aly Z, Shao JS, Lai CF, et al. Aortic Msx2-Wnt calcification cascade is regulated by TNF-α-dependent signals in diabetic Ldlr−/− mice. Arterioscler Thromb Vasc Biol. 2007;27(12):2589–96. doi: 10.1161/ATVBAHA.107.153668. [DOI] [PubMed] [Google Scholar]
  • 398.Wright WS, Longo KA, Dolinsky VW, et al. Wnt10b Inhibits Obesity in ob/ob and Agouti Mice. Diabetes. 2007;56(2):295–303. doi: 10.2337/db06-1339. [DOI] [PubMed] [Google Scholar]
  • 399.Lin CL, Wang JY, Huang YT, et al. Wnt/β-catenin signaling modulates survival of high glucose-stressed mesangial cells. J Am Soc Nephrol. 2006;17(10):2812–20. doi: 10.1681/ASN.2005121355. [DOI] [PubMed] [Google Scholar]
  • 400.Aslanidi G, Kroutov V, Philipsberg G, et al. Ectopic expression of Wnt10b decreases adiposity and improves glucose homeostasis in obese rats. Am J Physiol Endocrinol Metab. 2007;293(3):E726–36. doi: 10.1152/ajpendo.00248.2007. [DOI] [PubMed] [Google Scholar]
  • 401.Mani A, Radhakrishnan J, Wang H, et al. LRP6 mutation in a family with early coronary disease and metabolic risk factors. Science. 2007;315(5816):1278–82. doi: 10.1126/science.1136370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 402.Lynch RL, Konicek BW, McNulty AM, et al. The progression of LNCaP human prostate cancer cells to androgen independence involves decreased FOXO3a expression and reduced p27KIP1 promoter transactivation. Mol Cancer Res. 2005;3(3):163–9. doi: 10.1158/1541-7786.MCR-04-0163. [DOI] [PubMed] [Google Scholar]
  • 403.Hegde PS, Rusnak D, Bertiaux M, et al. Delineation of molecular mechanisms of sensitivity to lapatinib in breast cancer cell lines using global gene expression profiles. Mol Cancer Ther. 2007;6(5):1629–40. doi: 10.1158/1535-7163.MCT-05-0399. [DOI] [PubMed] [Google Scholar]
  • 404.Hoekstra AV, Ward EC, Hardt JL, et al. Chemosensitization of endometrial cancer cells through AKT inhibition involves FOXO1. Gynecol Oncol. 2008;108(3):609–18. doi: 10.1016/j.ygyno.2007.11.007. [DOI] [PubMed] [Google Scholar]
  • 405.Maiese K, Li F, Chong ZZ. Erythropoietin and cancer. JAMA. 2005;293(15):1858–9. doi: 10.1001/jama.293.15.1858-a. [DOI] [PubMed] [Google Scholar]
  • 406.Barandon L, Couffinhal T, Ezan J, et al. Reduction of infarct size and prevention of cardiac rupture in transgenic mice overexpressing FrzA. Circulation. 2003;108(18):2282–9. doi: 10.1161/01.CIR.0000093186.22847.4C. [DOI] [PubMed] [Google Scholar]
  • 407.Barandon L, Dufourcq P, Costet P, et al. Involvement of FrzA/sFRP-1 and the Wnt/frizzled pathway in ischemic preconditioning. Circ Res. 2005;96(12):1299–306. doi: 10.1161/01.RES.0000171895.06914.2c. [DOI] [PubMed] [Google Scholar]
  • 408.Emami KH, Corey E. When prostate cancer meets bone: control by wnts. Cancer Lett. 2007;253(2):170–9. doi: 10.1016/j.canlet.2006.12.040. [DOI] [PubMed] [Google Scholar]
  • 409.Sun J, Jin T. Both Wnt and mTOR signaling pathways are involved in insulin-stimulated proto-oncogene expression in intestinal cells. Cell Signal. 2008;20(1):219–29. doi: 10.1016/j.cellsig.2007.10.010. [DOI] [PubMed] [Google Scholar]

RESOURCES